1
|
Gómez GI, García-Rodríguez C, Marillán JE, Vergara SA, Alvear TF, Farias-Pasten A, Sáez JC, Retamal MA, Rovegno M, Ortiz FC, Orellana JA. Acute activation of hemichannels by ethanol leads to Ca 2+-dependent gliotransmitter release in astrocytes. Front Cell Dev Biol 2024; 12:1422978. [PMID: 38974144 PMCID: PMC11224458 DOI: 10.3389/fcell.2024.1422978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Multiple studies have demonstrated that acute ethanol consumption alters brain function and cognition. Nevertheless, the mechanisms underlying this phenomenon remain poorly understood. Astrocyte-mediated gliotransmission is crucial for hippocampal plasticity, and recently, the opening of hemichannels has been found to play a relevant role in this process. Hemichannels are plasma membrane channels composed of six connexins or seven pannexins, respectively, that oligomerize around a central pore. They serve as ionic and molecular exchange conduits between the cytoplasm and extracellular milieu, allowing the release of various paracrine substances, such as ATP, D-serine, and glutamate, and the entry of ions and other substances, such as Ca2+ and glucose. The persistent and exacerbated opening of hemichannels has been associated with the pathogenesis and progression of several brain diseases for at least three mechanisms. The uncontrolled activity of these channels could favor the collapse of ionic gradients and osmotic balance, the release of toxic levels of ATP or glutamate, cell swelling and plasma membrane breakdown and intracellular Ca2+ overload. Here, we evaluated whether acute ethanol exposure affects the activity of astrocyte hemichannels and the possible repercussions of this phenomenon on cytoplasmatic Ca2+ signaling and gliotransmitter release. Acute ethanol exposure triggered the rapid activation of connexin43 and pannexin1 hemichannels in astrocytes, as measured by time-lapse recordings of ethidium uptake. This heightened activity derived from a rapid rise in [Ca2+]i linked to extracellular Ca2+ influx and IP3-evoked Ca2+ release from intracellular Ca2+ stores. Relevantly, the acute ethanol-induced activation of hemichannels contributed to a persistent secondary increase in [Ca2+]i. The [Ca2+]i-dependent activation of hemichannels elicited by ethanol caused the increased release of ATP and glutamate in astroglial cultures and brain slices. Our findings offer fresh perspectives on the potential mechanisms behind acute alcohol-induced brain abnormalities and propose targeting connexin43 and pannexin1 hemichannels in astrocytes as a promising avenue to prevent deleterious consequences of alcohol consumption.
Collapse
Affiliation(s)
- Gonzalo I. Gómez
- Faculty of Health Sciences, Institute of Biomedical Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Claudia García-Rodríguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Jesús E. Marillán
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio A. Vergara
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tanhia F. Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Arantza Farias-Pasten
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C. Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Mauricio A. Retamal
- Programa de Comunicación Celular en Cancer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C. Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
2
|
Gómez GI, Alvear TF, Roa DA, Farias-Pasten A, Vergara SA, Mellado LA, Martinez-Araya CJ, Prieto-Villalobos J, García-Rodríguez C, Sánchez N, Sáez JC, Ortíz FC, Orellana JA. Cx43 hemichannels and panx1 channels contribute to ethanol-induced astrocyte dysfunction and damage. Biol Res 2024; 57:15. [PMID: 38576018 PMCID: PMC10996276 DOI: 10.1186/s40659-024-00493-2] [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: 12/23/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Alcohol, a widely abused drug, significantly diminishes life quality, causing chronic diseases and psychiatric issues, with severe health, societal, and economic repercussions. Previously, we demonstrated that non-voluntary alcohol consumption increases the opening of Cx43 hemichannels and Panx1 channels in astrocytes from adolescent rats. However, whether ethanol directly affects astroglial hemichannels and, if so, how this impacts the function and survival of astrocytes remains to be elucidated. RESULTS Clinically relevant concentrations of ethanol boost the opening of Cx43 hemichannels and Panx1 channels in mouse cortical astrocytes, resulting in the release of ATP and glutamate. The activation of these large-pore channels is dependent on Toll-like receptor 4, P2X7 receptors, IL-1β and TNF-α signaling, p38 mitogen-activated protein kinase, and inducible nitric oxide (NO) synthase. Notably, the ethanol-induced opening of Cx43 hemichannels and Panx1 channels leads to alterations in cytokine secretion, NO production, gliotransmitter release, and astrocyte reactivity, ultimately impacting survival. CONCLUSION Our study reveals a new mechanism by which ethanol impairs astrocyte function, involving the sequential stimulation of inflammatory pathways that further increase the opening of Cx43 hemichannels and Panx1 channels. We hypothesize that targeting astroglial hemichannels could be a promising pharmacological approach to preserve astrocyte function and synaptic plasticity during the progression of various alcohol use disorders.
Collapse
Affiliation(s)
- Gonzalo I Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Tanhia F Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Daniela A Roa
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Arantza Farias-Pasten
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Sergio A Vergara
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Luis A Mellado
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Claudio J Martinez-Araya
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Juan Prieto-Villalobos
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile
| | - Claudia García-Rodríguez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, 2360102, Chile
| | - Natalia Sánchez
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C Sáez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, 2360102, Chile
| | - Fernando C Ortíz
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Mechanisms of Myelin Formation and Repair Laboratory, Chacabuco 675, of. 212, Santiago, 8350347, Chile.
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile.
| |
Collapse
|
3
|
Harder EV, Franklin JP, VanRyzin JW, Reissner KJ. Astrocyte-Neuron Interactions in Substance Use Disorders. ADVANCES IN NEUROBIOLOGY 2024; 39:165-191. [PMID: 39190075 DOI: 10.1007/978-3-031-64839-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Engagement of astrocytes within the brain's reward circuitry has been apparent for approximately 30 years, when noncontingent drug administration was observed to lead to cytological markers of reactive astrocytes. Since that time, advanced approaches in rodent behavior and astrocyte monitoring have revealed complex interactions between astrocytes with drug type, animal sex, brain region, and dose and duration of drug administration. A number of studies now collectively reveal that rodent drug self-administration followed by prolonged abstinence results in decreased features of structure and synaptic colocalization of astrocytes. In addition, stimulation of astrocytes in the nucleus accumbens with DREADD receptors or pharmacological compounds opposes drug-seeking behavior. These findings provide a clear path for ongoing investigation into astrocytes as mediators of drug action in the brain and underscore the potential therapeutic utility of astrocytes in the regulation of drug craving and relapse vulnerability.
Collapse
Affiliation(s)
- Eden V Harder
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Janay P Franklin
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan W VanRyzin
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn J Reissner
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Munier J, Shen S, Rahal D, Hanna A, Marty V, O'Neill P, Fanselow M, Spigelman I. Chronic intermittent ethanol exposure disrupts stress-related tripartite communication to impact affect-related behavioral selection in male rats. Neurobiol Stress 2023; 24:100539. [PMID: 37131490 PMCID: PMC10149313 DOI: 10.1016/j.ynstr.2023.100539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/04/2023] Open
Abstract
Alcohol use disorder (AUD) is characterized by loss of intake control, increased anxiety, and susceptibility to relapse inducing stressors. Both astrocytes and neurons contribute to behavioral and hormonal consequences of chronic intermittent ethanol (CIE) exposure in animal models. Details on how CIE disrupts hypothalamic neuro-glial communication, which mediates stress responses are lacking. We conducted a behavioral battery (grooming, open field, reactivity to a single, uncued foot-shock, intermittent-access two-bottle choice ethanol drinking) followed by Ca2+ imaging in ex-vivo slices of paraventricular nucleus of the hypothalamus (PVN) from male rats exposed to CIE vapor or air-exposed controls. Ca2+ signals were evaluated in response to norepinephrine (NE) with or without selective α-adrenergic receptor (αAR) or GluN2B-containing N-methyl-D-aspartate receptor (NMDAR) antagonists, followed by dexamethasone (DEX) to mock a pharmacological stress response. Expectedly, CIE rats had altered anxiety-like, rearing, grooming, and drinking behaviors. Importantly, NE-mediated reductions in Ca2+ event frequency were blunted in both CIE neurons and astrocytes. Administration of the selective α1AR antagonist, prazosin, reversed this CIE-induced dysfunction in both cell types. Additionally, the pharmacological stress protocol reversed the altered basal Ca2+ signaling profile of CIE astrocytes. Signaling changes in astrocytes in response to NE were correlated with anxiety-like behaviors, such as the grooming:rearing ratio, suggesting tripartite synaptic function plays a role in switching between exploratory and stress-coping behavior. These data show how CIE exposure causes persistent changes to PVN neuro-glial function and provides the groundwork for how these physiological changes manifest in behavioral selection.
Collapse
Affiliation(s)
- J.J. Munier
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
- Corresponding author.
| | - S. Shen
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - D. Rahal
- Edna Bennett Pierce Prevention Research Center, The Pennsylvania State University, United States
| | - A. Hanna
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - V.N. Marty
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - P.R. O'Neill
- Hatos Center for Neuropharmacology, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, UCLA, United States
| | - M.S. Fanselow
- Department of Psychology, College of Life Sciences, Department of Psychiatry & Biobehavioral Science, David Geffen School of Medicine, UCLA, United States
| | - I. Spigelman
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
- Corresponding author. Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, 10833 Le Conte Avenue, 63-078 CHS, Los Angeles, CA, 90095-1668, United States.
| |
Collapse
|
5
|
Zhu G, Wang X, Chen L, Lenahan C, Fu Z, Fang Y, Yu W. Crosstalk Between the Oxidative Stress and Glia Cells After Stroke: From Mechanism to Therapies. Front Immunol 2022; 13:852416. [PMID: 35281064 PMCID: PMC8913707 DOI: 10.3389/fimmu.2022.852416] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Stroke is the second leading cause of global death and is characterized by high rates of mortality and disability. Oxidative stress is accompanied by other pathological processes that together lead to secondary brain damage in stroke. As the major component of the brain, glial cells play an important role in normal brain development and pathological injury processes. Multiple connections exist in the pathophysiological changes of reactive oxygen species (ROS) metabolism and glia cell activation. Astrocytes and microglia are rapidly activated after stroke, generating large amounts of ROS via mitochondrial and NADPH oxidase pathways, causing oxidative damage to the glial cells themselves and neurons. Meanwhile, ROS cause alterations in glial cell morphology and function, and mediate their role in pathological processes, such as neuroinflammation, excitotoxicity, and blood-brain barrier damage. In contrast, glial cells protect the Central Nervous System (CNS) from oxidative damage by synthesizing antioxidants and regulating the Nuclear factor E2-related factor 2 (Nrf2) pathway, among others. Although numerous previous studies have focused on the immune function of glial cells, little attention has been paid to the role of glial cells in oxidative stress. In this paper, we discuss the adverse consequences of ROS production and oxidative-antioxidant imbalance after stroke. In addition, we further describe the biological role of glial cells in oxidative stress after stroke, and we describe potential therapeutic tools based on glia cells.
Collapse
Affiliation(s)
- Ganggui Zhu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luxi Chen
- Department of Medical Genetics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Biomedical Science, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Zaixiang Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenhua Yu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Yildizhan K, Çinar R, Naziroğlu M. The involvement of TRPM2 on the MPP +-induced oxidative neurotoxicity and apoptosis in hippocampal neurons from neonatal mice: protective role of resveratrol. Neurol Res 2022; 44:636-644. [PMID: 35019826 DOI: 10.1080/01616412.2022.2027644] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is an age-related chronic neurodegenerative disease. Although PD is known to be a result of damage to hippocampal neurons, its molecular mechanism has yet to be completely clarified. The neurodegeneration in hippocampal neurons has been suggested to include excessive production of reactive oxygen species (ROS). Mitochondrial dysfunction and disruption of intracellular Ca2+ homeostasis play the most important role in the increase in ROS production for the cells. Remarkably, it is stated in the literature that especially the change of Ca2+ homeostasis triggers neuronal degeneration. TRPM2 is a unique calcium-permeable nonselective cation channel, and densest in the numberless neuronal population. The current study aims to elucidate the effect of antioxidant resveratrol (Resv) on TRPM2-mediated oxidative stress (OS) induced by 1-methyl-4-phenylpyridinium (MPP) exposure in the primary mouse hippocampal neurons. The neurons were divided into four groups as Control, Resv , MPP, and MPP+ Resv. In the current results, the activation of TRPM2 was observed in primary hippocampal neurons with MPP incubation. TRPM2 channel expression levels in the MPP group increased in hippocampal neurons after MPP exposure. In addition, intracellular free Ca2+ concentration and TRPM2 channel currents were highest in MPP groups, although they were decreased by the Resv treatment. In addition, mitochondrial membrane depolarization, ROS, caspase-3, caspase-9, and apoptosis values induced by MPP decreased with resveratrol treatment. In conclusion, in our study, we observed that the dysregulation of OS-induced TRPM2 channel activation in hippocampal neurons exposed to MPP caused apoptotic cell death in neurons, while the use of resveratrol had a protective effect by reducing OS resources in the environment.
Collapse
Affiliation(s)
- Kenan Yildizhan
- Department of Biophysics, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Ramazan Çinar
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Naziroğlu
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey.,Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.,Drug Discovery Unit, BSN Health, Analysis and Innovation Ltd. Inc. Teknokent, Isparta, Turkey
| |
Collapse
|
7
|
Gakare SG, Varghese SS, Patni PP, Wagh SA, Ugale RR. Prevention of glutamate excitotoxicity in lateral habenula alleviates ethanol withdrawal-induced somatic and behavioral effects in ethanol dependent mice. Behav Brain Res 2022; 416:113557. [PMID: 34453973 DOI: 10.1016/j.bbr.2021.113557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 12/22/2022]
Abstract
Ethanol withdrawal commonly leads to anxiety-related disorder, a central factor toward negative reinforcement leading to relapse. The lateral habenula (LHb), an epithalamic nucleus, has emerged to be critical for both reward and aversion processing. Recent studies have also implicated the hyperactivity of LHb, adding to the emergence of negative emotional states during withdrawal from addictive drugs. Herein, we have studied the effects of glutamate transporter inhibitor (PDC), GluN2B-containing NMDAR antagonist (Ro25-6981), and intracellular calcium chelator (BAPTA-AM) injection in LHb on ethanol withdrawal symptoms. We found that ethanol 4 g/kg 20 % w/v intragastric (i.g.) for 10 days followed by 24 h of withdrawal showed a significant increase in somatic signs characterized by vocalization, shaking, and scratching. It also increased locomotor activity and anxiety-like behavior, collectively showing expression of ethanol withdrawal symptoms. The intra-LHb administration of PDC (0.5 ng) worsened the effect of ethanol withdrawal, whereas Ro25-6981 (2 and 4 ng) and BAPTA-AM (6.5 and 13 ng) significantly reversed ethanol withdrawal-induced behavior evident by a decrease in somatic signs, locomotor activity, and anxiety-like behavior. Further, pretreatment of Ro25-6981 and BAPTA-AM reduced the neuronal loss, whereas PDC increased it compared to the vehicle-treated group, as evidenced by NeuN staining. Altogether, our results suggest that increased glutamate, GluN2B activation, and likely calcium increase indicative of glutamate excitotoxicity-induced neuronal loss in LHb possibly endorse the emergence of ethanol withdrawal symptoms, while their inhibition might help in alleviating the ethanol withdrawal symptoms.
Collapse
Affiliation(s)
- Sukanya G Gakare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Shejin S Varghese
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Paras P Patni
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Samruddhi A Wagh
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Rajesh R Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India.
| |
Collapse
|
8
|
Miguel-Hidalgo JJ. Astroglia in the Vulnerability and Maintenance of Alcohol Use Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:255-279. [PMID: 34888838 DOI: 10.1007/978-3-030-77375-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes induced in the morphology and the multiplicity of functional roles played by astrocytes in brain regions critical to the establishment and maintenance of alcohol abuse suggest that they make an important contribution to the vulnerability to alcohol use disorders. The understanding of the relevant mechanisms accounting for that contribution is complicated by the fact that alcohol itself acts directly on astrocytes altering their metabolism, gene expression, and plasticity, so that the ultimate result is a complex interaction of various cellular pathways, including intracellular calcium regulation, neuroimmune responses, and regulation of neurotransmitter and gliotransmitter release and uptake. The recent years have seen a steady increase in the characterization of several of the relevant mechanisms, but much remains to be done for a full understanding of the astrocytes' contribution to the vulnerability to alcohol dependence and abuse and for using that knowledge in designing effective therapies for AUDs.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
9
|
Cortical astrocytes regulate ethanol consumption and intoxication in mice. Neuropsychopharmacology 2021; 46:500-508. [PMID: 32464636 PMCID: PMC8027025 DOI: 10.1038/s41386-020-0721-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are fundamental building blocks of the central nervous system. Their dysfunction has been implicated in many psychiatric disorders, including alcohol use disorder, yet our understanding of their functional role in ethanol intoxication and consumption is very limited. Astrocytes regulate behavior through multiple intracellular signaling pathways, including G-protein coupled-receptor (GPCR)-mediated calcium signals. To test the hypothesis that GPCR-induced calcium signaling is also involved in the behavioral effects of ethanol, we expressed astrocyte-specific excitatory DREADDs in the prefrontal cortex (PFC) of mice. Activating Gq-GPCR signaling in PFC astrocytes increased drinking in ethanol-naïve mice, but not in mice with a history of ethanol drinking. In contrast, reducing calcium signaling with an astrocyte-specific calcium extruder reduced ethanol intake. Cortical astrocyte calcium signaling also altered the acute stimulatory and sedative-hypnotic effects of ethanol. Astrocyte-specific Gq-DREADD activation increased both the locomotor-activating effects of low dose ethanol and the sedative-hypnotic effects of a high dose, while reduced astrocyte calcium signaling diminished sensitivity to the hypnotic effects. In addition, we found that adenosine A1 receptors were required for astrocyte calcium activation to increase ethanol sedation. These results support integral roles for PFC astrocytes in the behavioral actions of ethanol that are due, at least in part, to adenosine receptor activation.
Collapse
|
10
|
Gonzalez A. Antioxidants and Neuron-Astrocyte Interplay in Brain Physiology: Melatonin, a Neighbor to Rely on. Neurochem Res 2021; 46:34-50. [PMID: 31989469 DOI: 10.1007/s11064-020-02972-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
This manuscript is a review focused onto the role of astrocytes in the protection of neurons against oxidative stress and how melatonin can contribute to the maintenance of brain homeostasis. The first part of the review is dedicated to the dependence of neurons on astrocytes by terms of survival under oxidative stress conditions. Additionally, the effects of melatonin against oxidative stress in the brain and its putative role in the protection against diseases affecting the brain are highlighted. The effects of melatonin on the physiology of neurons and astrocytes also are reviewed.
Collapse
Affiliation(s)
- Antonio Gonzalez
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, 10003, Cáceres, Spain.
| |
Collapse
|
11
|
Ye L, Orynbayev M, Zhu X, Lim EY, Dereddi RR, Agarwal A, Bergles DE, Bhat MA, Paukert M. Ethanol abolishes vigilance-dependent astroglia network activation in mice by inhibiting norepinephrine release. Nat Commun 2020; 11:6157. [PMID: 33268792 PMCID: PMC7710743 DOI: 10.1038/s41467-020-19475-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Norepinephrine adjusts sensory processing in cortical networks and gates plasticity enabling adaptive behavior. The actions of norepinephrine are profoundly altered by recreational drugs like ethanol, but the consequences of these changes on distinct targets such as astrocytes, which exhibit norepinephrine-dependent Ca2+ elevations during vigilance, are not well understood. Using in vivo two-photon imaging, we show that locomotion-induced Ca2+ elevations in mouse astroglia are profoundly inhibited by ethanol, an effect that can be reversed by enhancing norepinephrine release. Vigilance-dependent astroglial activation is abolished by deletion of α1A-adrenergic receptor from astroglia, indicating that norepinephrine acts directly on these ubiquitous glial cells. Ethanol reduces vigilance-dependent Ca2+ transients in noradrenergic terminals, but has little effect on astroglial responsiveness to norepinephrine, suggesting that ethanol suppresses their activation by inhibiting norepinephrine release. Since abolition of astroglia Ca2+ activation does not affect motor coordination, global suppression of astroglial networks may contribute to the cognitive effects of alcohol intoxication. The effects of norepinephrine on sensory processing in cortical networks are altered by recreational drugs like ethanol. The authors show that ethanol suppresses the activation of astrocytes by inhibiting norepinephrine release which may contribute to the cognitive effects of alcohol intoxication.
Collapse
Affiliation(s)
- Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Murat Orynbayev
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiangyu Zhu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Eunice Y Lim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ram R Dereddi
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
12
|
Chen Y, Qin C, Huang J, Tang X, Liu C, Huang K, Xu J, Guo G, Tong A, Zhou L. The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif 2020; 53:e12781. [PMID: 32035016 PMCID: PMC7106951 DOI: 10.1111/cpr.12781] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/17/2019] [Accepted: 01/20/2020] [Indexed: 02/05/2023] Open
Abstract
Central nervous system (CNS) maintains a high level of metabolism, which leads to the generation of large amounts of free radicals, and it is also one of the most vulnerable organs to oxidative stress. Emerging evidences have shown that, as the key homeostatic cells in CNS, astrocytes are deeply involved in multiple aspects of CNS function including oxidative stress regulation. Besides, the redox level in CNS can in turn affect astrocytes in morphology and function. The complex and multiple roles of astrocytes indicate that their correct performance is crucial for the normal functioning of the CNS, and its dysfunction may result in the occurrence and progression of various neurological disorders. To date, the influence of astrocytes in CNS oxidative stress is rarely reviewed. Therefore, in this review we sum up the roles of astrocytes in redox regulation and the corresponding mechanisms under both normal and different pathological conditions.
Collapse
Affiliation(s)
- Yaxing Chen
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Qin
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Jianhan Huang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Tang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Keru Huang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Guo
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
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: 159] [Impact Index Per Article: 22.7] [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
|
14
|
Orellana JA, Cerpa W, Carvajal MF, Lerma-Cabrera JM, Karahanian E, Osorio-Fuentealba C, Quintanilla RA. New Implications for the Melanocortin System in Alcohol Drinking Behavior in Adolescents: The Glial Dysfunction Hypothesis. Front Cell Neurosci 2017; 11:90. [PMID: 28424592 PMCID: PMC5380733 DOI: 10.3389/fncel.2017.00090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Alcohol dependence causes physical, social, and moral harms and currently represents an important public health concern. According to the World Health Organization (WHO), alcoholism is the third leading cause of death worldwide, after tobacco consumption and hypertension. Recent epidemiologic studies have shown a growing trend in alcohol abuse among adolescents, characterized by the consumption of large doses of alcohol over a short time period. Since brain development is an ongoing process during adolescence, short- and long-term brain damage associated with drinking behavior could lead to serious consequences for health and wellbeing. Accumulating evidence indicates that alcohol impairs the function of different components of the melanocortin system, a major player involved in the consolidation of addictive behaviors during adolescence and adulthood. Here, we hypothesize the possible implications of melanocortins and glial cells in the onset and progression of alcohol addiction. In particular, we propose that alcohol-induced decrease in α-MSH levels may trigger a cascade of glial inflammatory pathways that culminate in altered gliotransmission in the ventral tegmental area and nucleus accumbens (NAc). The latter might potentiate dopaminergic drive in the NAc, contributing to increase the vulnerability to alcohol dependence and addiction in the adolescence and adulthood.
Collapse
Affiliation(s)
- Juan A Orellana
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Laboratorio de Neurociencias, Departamento de Neurología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Waldo Cerpa
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Maria F Carvajal
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Unidad de Neurociencia, Centro de Investigación Biomédica, Universidad Autónoma de ChileSantiago, Chile
| | - José M Lerma-Cabrera
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Unidad de Neurociencia, Centro de Investigación Biomédica, Universidad Autónoma de ChileSantiago, Chile
| | - Eduardo Karahanian
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Unidad de Neurociencia, Centro de Investigación Biomédica, Universidad Autónoma de ChileSantiago, Chile
| | - Cesar Osorio-Fuentealba
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Facultad de Kinesiología, Artes y Educación Física, Universidad Metropolitana de Ciencias de la EducaciónSantiago, Chile
| | - Rodrigo A Quintanilla
- Centro de Investigación y Estudio del Consumo de Alcohol en AdolescentesSantiago, Chile.,Laboratory of Neurodegenerative Diseases, Universidad Autónoma de ChileSantiago, Chile
| |
Collapse
|
15
|
Carter JM, Landin JD, Gigante ED, Rieger SP, Diaz MR, Werner DF. Inhibitors of Calcium-Activated Anion Channels Modulate Hypnotic Ethanol Responses in Adult Sprague Dawley Rats. Alcohol Clin Exp Res 2016; 40:301-8. [PMID: 26842249 DOI: 10.1111/acer.12957] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 11/02/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ethanol is widely known for its depressant effects; however, the underlying neurobiological mechanisms are not clear. Calcium-activated anion channels (CAACs) contribute to extracellular chloride levels and thus may be involved in regulating inhibitory mechanisms within the central nervous system. Therefore, we hypothesized that CAACs influence ethanol behavioral sensitivity by altering CAAC expression. METHODS We assessed the role of CAACs in ethanol-induced loss of righting reflex (LORR) and locomotor activity using intracerebroventricular infusions of several nonselective CAAC blockers. CAAC expression was determined after ethanol exposure. RESULTS Ethanol-induced LORR (4.0 g/kg, intraperitoneally [i.p.]) was significantly attenuated by all 4 CAAC blockers. Blocking CAACs did not impact ethanol's low-dose (1.5 g/kg, i.p.) locomotor-impairing effects. Biochemical analysis of CAAC protein expression revealed that cortical Bestrophin1 (Best1) and Tweety1 levels were reduced as early as 30 minutes following a single ethanol injection (3.5 g/kg, intraperitoneally [i.p.]) and remained decreased 24 hours later in P2 fractions. Cortical Best1 levels were also reduced following 1.5 g/kg. However, CAAC expression was unaltered in the striatum following a single ethanol exposure. Ethanol did not affect Tweety2 levels in either brain region. CONCLUSIONS These results suggest that CAACs are a major target of ethanol in vivo, and the regulation of these channels contributes to select behavioral actions of ethanol.
Collapse
Affiliation(s)
- Jenna M Carter
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, New York
| | - Justine D Landin
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, New York
| | - Eduardo D Gigante
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, New York.,Department of Health and Human Services, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - Samuel P Rieger
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, New York
| | - Marvin R Diaz
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, New York
| | - David F Werner
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, New York
| |
Collapse
|
16
|
Adermark L, Bowers MS. Disentangling the Role of Astrocytes in Alcohol Use Disorder. Alcohol Clin Exp Res 2016; 40:1802-16. [PMID: 27476876 PMCID: PMC5407469 DOI: 10.1111/acer.13168] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/02/2016] [Indexed: 01/29/2023]
Abstract
Several laboratories recently identified that astrocytes are critical regulators of addiction machinery. It is now known that astrocyte pathology is a common feature of ethanol (EtOH) exposure in both humans and animal models, as even brief EtOH exposure is sufficient to elicit long-lasting perturbations in astrocyte gene expression, activity, and proliferation. Astrocytes were also recently shown to modulate the motivational properties of EtOH and other strongly reinforcing stimuli. Given the role of astrocytes in regulating glutamate homeostasis, a crucial component of alcohol use disorder (AUD), astrocytes might be an important target for the development of next-generation alcoholism treatments. This review will outline some of the more prominent features displayed by astrocytes, how these properties are influenced by acute and long-term EtOH exposure, and future directions that may help to disentangle astrocytic from neuronal functions in the etiology of AUD.
Collapse
Affiliation(s)
- Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Box 410, SE-405 30 Gothenburg, Sweden
| | - M. Scott Bowers
- Department of Psychiatry, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Faulk Center for Molecular Therapeutics, Northwestern University; Aptinyx,, Evanston, Il 60201, USA
| |
Collapse
|
17
|
Harada K, Kamiya T, Tsuboi T. Gliotransmitter Release from Astrocytes: Functional, Developmental, and Pathological Implications in the Brain. Front Neurosci 2016; 9:499. [PMID: 26793048 PMCID: PMC4709856 DOI: 10.3389/fnins.2015.00499] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022] Open
Abstract
Astrocytes comprise a large population of cells in the brain and are important partners to neighboring neurons, vascular cells, and other glial cells. Astrocytes not only form a scaffold for other cells, but also extend foot processes around the capillaries to maintain the blood–brain barrier. Thus, environmental chemicals that exist in the blood stream could have potentially harmful effects on the physiological function of astrocytes. Although astrocytes are not electrically excitable, they have been shown to function as active participants in the development of neural circuits and synaptic activity. Astrocytes respond to neurotransmitters and contribute to synaptic information processing by releasing chemical transmitters called “gliotransmitters.” State-of-the-art optical imaging techniques enable us to clarify how neurotransmitters elicit the release of various gliotransmitters, including glutamate, D-serine, and ATP. Moreover, recent studies have demonstrated that the disruption of gliotransmission results in neuronal dysfunction and abnormal behaviors in animal models. In this review, we focus on the latest technical approaches to clarify the molecular mechanisms of gliotransmitter exocytosis, and discuss the possibility that exposure to environmental chemicals could alter gliotransmission and cause neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| | - Taichi Kamiya
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| |
Collapse
|
18
|
Wilhelm CJ, Hashimoto JG, Roberts ML, Bloom SH, Andrew MR, Wiren KM. Astrocyte Dysfunction Induced by Alcohol in Females but Not Males. Brain Pathol 2015; 26:433-51. [PMID: 26088166 DOI: 10.1111/bpa.12276] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/10/2015] [Indexed: 01/08/2023] Open
Abstract
Chronic alcohol abuse is associated with brain damage in a sex-specific fashion, but the mechanisms involved are poorly described and remain controversial. Previous results have suggested that astrocyte gene expression is influenced by ethanol intoxication and during abstinence in vivo. Here, bioinformatic analysis of astrocyte-enriched ethanol-regulated genes in vivo revealed ubiquitin pathways as an ethanol target, but with sexually dimorphic cytokine signaling and changes associated with brain aging in females and not males. Consistent with this result, astrocyte activation was observed after exposure in female but not male animals, with reduced S100β levels in the anterior cingulate cortex and increased GFAP(+) cells in the hippocampus. In primary culture, the direct effects of chronic ethanol exposure followed by recovery on sex-specific astrocyte function were examined. Male astrocyte responses were consistent with astrocyte deactivation with reduced GFAP expression during ethanol exposure. In contrast, female astrocytes exhibited increased expression of Tnf, reduced expression of the neuroprotective cytokine Tgfb1, disrupted bioenergetics and reduced excitatory amino acid uptake following exposure or recovery. These results indicate widespread astrocyte dysfunction in ethanol-exposed females and suggest a mechanism that may underlie increased vulnerability to ethanol-induced neurotoxicity in females.
Collapse
Affiliation(s)
- Clare J Wilhelm
- VA Portland Health Care System, Portland, OR.,Department of Psychiatry, Oregon Health & Science University, Portland, OR
| | - Joel G Hashimoto
- VA Portland Health Care System, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | | | | | - Melissa R Andrew
- Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kristine M Wiren
- VA Portland Health Care System, Portland, OR.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| |
Collapse
|
19
|
Baliño P, Ledesma JC, Aragon CMG. Role of CA2+/calmodulin on ethanol neurobehavioral effects. Psychopharmacology (Berl) 2014; 231:4611-21. [PMID: 24853690 DOI: 10.1007/s00213-014-3610-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 04/29/2014] [Indexed: 01/13/2023]
Abstract
RATIONALE The cAMP-dependent protein kinase A (PKA) signaling transduction pathway has been shown to play an important role in the modulation of several ethanol-induced behaviors. Different studies have demonstrated intracellular calcium (Ca(2+))-dependent activation of the PKA cascade after ethanol administration. Thus, the cAMP cascade mediator Ca(2+)-dependent calmodulin (CaM) has been strongly implicated in the central effects of ethanol. OBJECTIVES In this study, we assessed the role of the CaM inhibitor W7 on ethanol-induced stimulation, ethanol intake, and ethanol-induced activation of PKA. METHODS Swiss mice were pretreated with W7 (0-10 mg/kg) 30 min before ethanol (0-3.75 g/kg) administration. Immediately, animals were placed during 20 min in an open-field chamber. Ethanol (10 %, v/v) intake in 2 h was assessed using a limited access paradigm. Experiments with caffeine (0-15 mg/kg), cocaine (0-4 mg/kg), and saccharine (0.1 %, w/v) were designed to compare their results to those obtained with ethanol. Western blot was assayed 45 min after ethanol administration. RESULTS Results showed that pretreatment with W7, reduced selectively in a dose-dependent fashion ethanol-induced locomotor stimulation and ethanol intake. The ethanol-induced activation of PKA was also prevented by W7 administration. CONCLUSIONS These results demonstrate that CaM inhibition resulted in a selective reduction of ethanol-stimulating effects and ethanol intake. The PKA activation induced by ethanol was blocked after the CaM blockade with W7. These results provide further evidence of the key role of cellular Ca(2+)-dependent pathways on the central effects of ethanol.
Collapse
Affiliation(s)
- Pablo Baliño
- Área de Psicobiología, Universtitat Jaume I, Avda. Sos Baynat s/n, 12071, Castellón, Spain,
| | | | | |
Collapse
|
20
|
Baliño P, Ledesma JC, Aragon CMG. In vivo study of ethanol-activated brain protein kinase A: manipulations of Ca2+ distribution and flux. Alcohol Clin Exp Res 2013; 38:629-40. [PMID: 24117724 DOI: 10.1111/acer.12289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/21/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND The cAMP-dependent protein kinase (PKA) signaling transduction pathway has been shown to play an important role in the modulation of several ethanol (EtOH)-induced behavioral actions. In vivo, short-term exposure to EtOH up-regulates the cAMP-signaling cascade. Interestingly, different Ca(2+) -dependent cAMP-PKA cascade mediators play a critical role in the neurobehavioral response to EtOH, being of special relevance to the Ca(2+) -dependent adenylyl cyclases 1 and 8. We hypothesize an intracellular PKA activation elicited by EtOH administration, which may be regulated by a Ca(2+) -dependent mechanism as an early cellular response. Thus, the present work aims to explore the role of Ca(2+) (internal and external) on the EtOH-activated PKA cascade. METHODS Swiss male mice received an intraperitoneal injection of EtOH (0 or 4 g/kg), and brains were dissected following a temporal pattern (7, 15, 30, 45, 90, or 120 minutes). Either the enzymatic PKA activity or its fingerprint was analyzed on different brain areas (cortex, hypothalamus, hippocampus, and striatum). To explore the role of Ca(2+) on the EtOH-activated PKA cascade, mice were pretreated with diltiazem (0 or 20 mg/kg), dantrolene (0 or 5 mg/kg), or 3,7-Dimethyl-1-(2-propynyl)xanthine (0 or 1 mg/kg) 30 minutes before EtOH (4 g/kg) administration. After 45 minutes of EtOH administration, brains were removed and dissected to measure the PKA activity or its fingerprint. RESULTS Results from these experiments showed an EtOH-dependent activation of PKA in different brain areas. Manipulations involving a disruption of intracellular Ca(2+) release from the endoplasmic reticulum resulted in a decreased EtOH-induced activation of PKA. On the contrary, extracellular-to-cytoplasm Ca(2+) manipulations did not prevent the PKA activation by EtOH. CONCLUSIONS Altogether, these results show the critical role of stored Ca(2+) as an intracellular mediator of different neurobiological actions of EtOH and provide further evidence of a possible new target for EtOH within the central nervous system.
Collapse
Affiliation(s)
- Pablo Baliño
- Area de Psicobiologia, Universitat Jaume I, Castellón, Spain
| | | | | |
Collapse
|
21
|
Wiren KM. Males and females are just different: Sexually dimorphic responses to chronic ethanol exposure in hippocampal slice cultures. Neurosci Lett 2013; 550:1-5. [DOI: 10.1016/j.neulet.2013.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 01/07/2023]
|
22
|
Yuan H, Zhang J, Liu H, Li Z. The protective effects of resveratrol on Schwann cells with toxicity induced by ethanol in vitro. Neurochem Int 2013; 63:146-53. [PMID: 23770283 DOI: 10.1016/j.neuint.2013.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/27/2013] [Accepted: 05/26/2013] [Indexed: 10/26/2022]
Abstract
Schwann cells (SCs) are the myelin forming cells in the peripheral nervous system, they play a key role in the pathology of various polyneuropathies and provide trophic support to axons via expression of various neurotrophic factors, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). Ethanol (EtOH) adversely affected both SCs proliferation and myelin formation in culture. Resveratrol (Res) has been shown to regulate many cellular processes and to display multiple protective and therapeutic effects. Whether Res has protective effects on SCs with EtOH-induced toxicity is still unclear. The protective efficacy of Res on EtOH-treated SCs in vitro was investigated in the present study. Res improved cell viability of the EtOH-treated SCs. Hoechst 33342 staining and terminal deoxynucleotidyl transferase (TdT)-mediated deoxyuridine triphosphate nick-end labeling analysis showed that the EtOH-induced apoptosis was inhibited by Res. The effects of Res were blocked by the 5'-adenosine monophosphate-activated protein kinase inhibitor Compound C and the silencing information regulator T1 inhibitor nicotinamide. Res could increase the mRNA and protein levels of BDNF and GDNF in the EtOH-treated SCs. However, the EtOH-induced increase of NGF in the SCs is inhibited by Res. The data from the present study indicate that Res protects SCs from EtOH-induced cell death and regulates the expression of neurotrophicfactors. Res and its derivative may be effective for the treatment of neuropathic diseases induced by EtOH.
Collapse
Affiliation(s)
- Hongtu Yuan
- Department of Anatomy, Shandong University School of Medicine, Jinan 250012, China.
| | | | | | | |
Collapse
|
23
|
Hutchinson MR, Watkins LR. Why is neuroimmunopharmacology crucial for the future of addiction research? Neuropharmacology 2013; 76 Pt B:218-27. [PMID: 23764149 DOI: 10.1016/j.neuropharm.2013.05.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/13/2013] [Accepted: 05/23/2013] [Indexed: 12/13/2022]
Abstract
A major development in drug addiction research in recent years has been the discovery that immune signaling within the central nervous system contributes significantly to mesolimbic dopamine reward signaling induced by drugs of abuse, and hence is involved in the presentation of reward behaviors. Additionally, in the case of opioids, these hypotheses have advanced through to the discovery of the novel site of opioid action at the innate immune pattern recognition receptor Toll-like receptor 4 as the necessary triggering event that engages this reward facilitating central immune signaling. Thus, the hypothesis of major proinflammatory contributions to drug abuse was born. This review will examine these key discoveries, but also address several key lingering questions of how central immune signaling is able to contribute in this fashion to the pharmacodynamics of drugs of abuse. It is hoped that by combining the collective wisdom of neuroscience, immunology and pharmacology, into Neuroimmunopharmacology, we may more fully understanding the neuronal and immune complexities of how drugs of abuse, such as opioids, create their rewarding and addiction states. Such discoveries will point us in the direction such that one day soon we might successfully intervene to successfully treat drug addiction. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Level 5, Medical School South, Frome Rd, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
24
|
Rakofsky JJ, Dunlop BW. Do alcohol use disorders destabilize the course of bipolar disorder? J Affect Disord 2013; 145:1-10. [PMID: 22858208 DOI: 10.1016/j.jad.2012.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/12/2012] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To determine whether long-term data implicate a negative effect of alcohol-use disorders (AUDs) on time to remission, risk of mood episode recurrence, and risk of mood switch/cycling in patients with bipolar disorder (BD). The short-term temporal sequence between alcohol use and onset of mood episodes was also examined. METHODS A MEDLINE literature search was conducted for measurement-based reports of alcohol and course of bipolar disorder. RESULTS Twenty-three original data publications were identified. Three out of 5 studies addressing the impact of AUDs on recovery from a mood episode demonstrated that alcohol did not prolong index mood episodes of any type. Six out of 11 reports evaluating the relationship between alcohol and the long term risk of mood episode recurrences suggested that high levels of alcohol intake increase the risk of a mood recurrence. Five out of 7 studies evaluating the short-term temporal sequence of AUDs and development of mood episodes among BD patients found that increased alcohol use preceded the development of new mood episodes. Four out of 5 studies examining the association between alcohol and rapid cycling indicated that AUDs were associated with higher rates of rapid-cycling. LIMITATIONS We limited our review to studies that were large enough to perform statistical testing, which may have led us to overlook informative smaller studies. CONCLUSIONS Although alcohol does not seem to affect time to mood episode remission, alcohol use destabilizes the course of illness over the long run as evidenced by associations with more rapid cycling and mood episode recurrence.
Collapse
Affiliation(s)
- Jeffrey J Rakofsky
- Mood and Anxiety Disorders Program, Emory University, Department of Psychiatry and Behavioral Sciences, 1256 Briarcliff Rd, 3rd Floor North, Atlanta, GA 30306, USA.
| | | |
Collapse
|
25
|
Tarragon E, Baliño P, Aragon CMG. Dantrolene blockade of ryanodine receptor impairs ethanol-induced behavioral stimulation, ethanol intake and loss of righting reflex. Behav Brain Res 2012; 233:554-62. [PMID: 22677274 DOI: 10.1016/j.bbr.2012.05.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 05/22/2012] [Accepted: 05/26/2012] [Indexed: 12/21/2022]
Abstract
Calcium has been characterized as one of the most ubiquitous, universal and versatile intracellular signals. Among other substances with the ability to alter intracellular calcium levels, ethanol has been described as particularly relevant because of its social and economic impact. Ethanol effects on calcium distribution and flux in vitro have been widely studied, showing that acute ethanol administration can modulate intracellular calcium concentrations in a dose dependent manner. Intracellular calcium released from the endoplasmic reticulum plays a determinant role in several cellular processes. In this study, we aim to assess the effect of dantrolene, a ryanodine receptor antagonist, on three different ethanol-elicited behaviors: locomotor activity, loss of righting reflex and ethanol intake. Mice were challenged with an injection of dantrolene (0-5 mg/kg, i.p.) 30 min before ethanol (0-4 g/kg, i.p.) administration. Animals were immediately placed in an open field cylinder to monitor distance travelled horizontally or in a V-shaped trough to measure righting reflex recovery time. For ethanol intake, dantrolene (0-5mg/kg, i.p.) was administered 30 min before ethanol (20%, v/v) exposure, following a drinking in the dark paradigm. Our results showed that dantrolene selectively reduces ethanol-induced stimulation, loss of righting reflex, and ethanol intake in a dose dependent manner. Together, these data suggest that intracellular calcium released from the endoplasmic reticulum may play a critical role in behavioral effects caused by ethanol, and point to a calcium-dependent pathway as a possible cellular mechanism of action for ethanol.
Collapse
Affiliation(s)
- Ernesto Tarragon
- Area de Psicobiologia, Universitat Jaume I, 12071 Castellón, Spain
| | | | | |
Collapse
|
26
|
Coller JK, Hutchinson MR. Implications of central immune signaling caused by drugs of abuse: mechanisms, mediators and new therapeutic approaches for prediction and treatment of drug dependence. Pharmacol Ther 2012; 134:219-45. [PMID: 22316499 DOI: 10.1016/j.pharmthera.2012.01.008] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 01/12/2023]
Abstract
In the past two decades a trickle of manuscripts examining the non-neuronal central nervous system immune consequences of the drugs of abuse has now swollen to a significant body of work. Initially, these studies reported associative evidence of central nervous system proinflammation resulting from exposure to the drugs of abuse demonstrating key implications for neurotoxicity and disease progression associated with, for example, HIV infection. However, more recently this drug-induced activation of central immune signaling is now understood to contribute substantially to the pharmacodynamic actions of the drugs of abuse, by enhancing the engagement of classical mesolimbic dopamine reward pathways and withdrawal centers. This review will highlight the key in vivo animal, human, biological and molecular evidence of these central immune signaling actions of opioids, alcohol, cocaine, methamphetamine, and 3,4-methylenedioxymethamphetamine (MDMA). Excitingly, this new appreciation of central immune signaling activity of drugs of abuse provides novel therapeutic interventions and opportunities to identify 'at risk' individuals through the use of immunogenetics. Discussion will also cover the evidence of modulation of this signaling by existing clinical and pre-clinical drug candidates, and novel pharmacological targets. Finally, following examination of the breadth of central immune signaling actions of the drugs of abuse highlighted here, the current known common immune signaling components will be outlined and their impact on established addiction neurocircuitry discussed, thereby synthesizing a common neuroimmune hypothesis of addiction.
Collapse
Affiliation(s)
- Janet K Coller
- Discipline of Pharmacology, School of Medical Sciences, University of Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
27
|
Intracellular calcium chelation with BAPTA-AM modulates ethanol-induced behavioral effects in mice. Exp Neurol 2012; 234:446-53. [PMID: 22306018 DOI: 10.1016/j.expneurol.2012.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 01/11/2012] [Accepted: 01/16/2012] [Indexed: 11/23/2022]
Abstract
Calcium (Ca(2+)) has been characterized as one of the most ubiquitous, universal and versatile intracellular signaling molecules responsible for controlling numerous cellular processes. Ethanol-induced effects on Ca(2+) distribution and flux have been widely studied in vitro, showing that acute ethanol administration can modulate intracellular Ca(2+) concentrations in a dose dependent manner. In vivo, the relationship between Ca(2+) manipulation and the corresponding ethanol-induced behavioral effects have focused on Ca(2+) flux through voltage-gated Ca(2+) channels. The present study investigated the role of inward Ca(2+) currents in ethanol-induced psychomotor effects (stimulation and sedation) and ethanol intake. We studied the effects of the fast Ca(2+) chelator, BAPTA-AM, on ethanol-induced locomotor activity and the sedative effects of ethanol. Swiss (RjOrl) mice were pretreated with BAPTA-AM (0-10 mg/kg) 30 min before an ethanol (0-4 g/kg) challenge. Our results revealed that pretreatment with BAPTA-AM prevented locomotor stimulation produced by ethanol without altering basal locomotion. In contrast, BAPTA-AM reversed ethanol-induced hypnotic effects. In a second set of experiments, we investigated the effects of intracellular Ca(2+) chelation on ethanol intake. Following a drinking-in-the-dark methodology, male C57BL/6J mice were offered 20% v/v ethanol, tap water, or 0.1% sweetened water. The results of these experiments revealed that BAPTA-AM pretreatment (0-5 mg/kg) reduced ethanol consumption in a dose-dependent manner while leaving water and sweetened water intake unaffected. Our findings support the role of inward Ca(2+) currents in mediating different behavioral responses induced by ethanol. Our results are discussed together with data indicating that ethanol appears to be more sensitive to intracellular Ca(2+) manipulations than other psychoactive drugs.
Collapse
|
28
|
Zorumski CF, Izumi Y. NMDA receptors and metaplasticity: mechanisms and possible roles in neuropsychiatric disorders. Neurosci Biobehav Rev 2012; 36:989-1000. [PMID: 22230702 DOI: 10.1016/j.neubiorev.2011.12.011] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 11/14/2011] [Accepted: 12/22/2011] [Indexed: 02/08/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are key components of neural signaling, playing roles in synaptic transmission and in the synaptic plasticity thought to underlie learning and memory. NMDAR activation can also have neurotoxic consequences contributing to several forms of neurodegeneration. Additionally, NMDARs can modulate neuronal function and regulate the ability of synapses to undergo synaptic plasticity. Evidence gathered over the past 20 years strongly supports the idea that untimely activation of NMDARs impairs the induction of long-term potentiation (LTP) by a form of metaplasticity. This metaplasticity can be triggered by multiple stimuli including physiological receptor activation, and metabolic and behavioral stressors. These latter findings raise the possibility that NMDARs contribute to cognitive dysfunction associated with neuropsychiatric disorders. This paper examines NMDAR metaplasticity and its potential role in cognition. Recent studies using NMDAR antagonists for therapeutic purposes also raise the possibility that metaplasticity may contribute to clinical effects of certain drugs.
Collapse
Affiliation(s)
- Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | |
Collapse
|
29
|
Gruol DL, Nelson TE, Hao C, Michael S, Vukojevic V, Ming Y, Terenius L. Ethanol alters opioid regulation of Ca(2+) influx through L-type Ca(2+) channels in PC12 cells. Alcohol Clin Exp Res 2011; 36:443-56. [PMID: 22014285 DOI: 10.1111/j.1530-0277.2011.01631.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Studies at the behavioral and synaptic level show that effects of ethanol on the central nervous system can involve the opioid signaling system. These interactions may alter the function of a common downstream target. In this study, we examined Ca(2+) channel function as a potential downstream target of interactions between ethanol and μ or κ opioid receptor signaling. METHODS The studies were carried out in a model system, undifferentiated PC12 cells transfected with μ or κ opioid receptors. The PC12 cells express L-type Ca(2+) channels, which were activated by K(+) depolarization. Ca(2+) imaging was used to measure relative Ca(2+) flux during K(+) depolarization and the modulation of Ca(2+) flux by opioids and ethanol. RESULTS Ethanol, μ receptor activation, and κ receptor activation all reduced the amplitude of the Ca(2+) signal produced by K(+) depolarization. Pretreatment with ethanol or combined treatment with ethanol and μ or κ receptor agonists caused a reduction in the amplitude of the Ca(2+) signal that was comparable to or smaller than that observed for the individual drugs alone, indicating an interaction by the drugs at a downstream target (or targets) that limited the modulation of Ca(2+) flux through L-type Ca(2+) channels. CONCLUSIONS These studies provide evidence for a cellular mechanism that could play an important role in ethanol regulation of synaptic transmission and behavior through interactions with the opioid signaling.
Collapse
Affiliation(s)
- Donna L Gruol
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Collins MA, Neafsey EJ. Neuroinflammatory pathways in binge alcohol-induced neuronal degeneration: oxidative stress cascade involving aquaporin, brain edema, and phospholipase A2 activation. Neurotox Res 2011; 21:70-8. [PMID: 21927955 DOI: 10.1007/s12640-011-9276-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 09/01/2011] [Accepted: 09/05/2011] [Indexed: 12/21/2022]
Abstract
Chronic binge alcohol exposure in adult rat models causes neuronal degeneration in the cortex and hippocampus that is not reduced by excitotoxic receptor antagonists, but is prevented by antioxidants. Neuroinflammatory (glial-neuronal) signaling pathways are believed to underlie the oxidative stress and brain damage. Based on our experimental results as well as increased knowledge about the pro-neuroinflammatory potential of glial water channels, we propose that induction of aquaporin-4 can be a critical initiating factor in alcohol's neurotoxic effects, through the instigation of cellular edema-based neuroinflammatory cascades involving increased phospholipase A2 activities, polyunsaturated fatty acid release/membrane depletion, decreased prosurvival signaling, and oxidative stress. A testable scheme for this pathway is presented that incorporates recent findings in the alcohol-brain literature indicating a role for neuroimmune activation (upregulation of NF-kappaB, proinflammatory cytokines, and toll-like receptors). We present the argument that such neuroimmune activation could be associated with or even dependent on increased aquaporin-4 and glial swelling as well.
Collapse
Affiliation(s)
- Michael A Collins
- Department of Molecular Pharmacology & Therapeutics, Stritch School of Medicine, Loyola University Chicago, 2160 South First Avenue, Maywood, IL 60153, USA.
| | | |
Collapse
|
31
|
Santofimia-Castaño P, Salido GM, Gonzalez A. Ethanol reduces kainate-evoked glutamate secretion in rat hippocampal astrocytes. Brain Res 2011; 1402:1-8. [PMID: 21679931 DOI: 10.1016/j.brainres.2011.05.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 05/25/2011] [Accepted: 05/25/2011] [Indexed: 12/12/2022]
Abstract
In this study we have used rat hippocampal astrocytes in culture to investigate the effect of ethanol on kainate-induced glutamate secretion. Our results show that kainate (10 μM to 500 μM) stimulated glutamate release from astrocytes. Preincubation of astrocytes in the presence of ethanol induced a concentration-dependent (1mM-50mM) inhibition of glutamate release caused by stimulation of cells with 100 μM kainate. Inhibition of alcohol-dehydrogenase, by preincubation of astrocytes in the presence of 4-methylpyrazole (1mM), abolished ethanol-induced inhibition of glutamate release in response to kainate. On the other hand, preincubation of astrocytes in the presence of the antioxidant cinnamtannin B-1 (10 μM) also blocked ethanol inhibitory action on glutamate release in response to kainate. Ethanol (50mM) reduced Ca(2+) mobilization in response to kainate, whereas cinnamtannin B-1 reversed the inhibitory action of ethanol on Ca(2+) mobilization by kainate. Our results are consistent with an inhibitory action of ethanol on glutamate secretion from hippocampal astrocytes. The inhibitory effects of ethanol are probably due to its oxidative metabolization, involves reactive oxygen species production, and a lower Ca(2+) mobilization by kainate. Taking into account the pivotal role that astrocytes play within the central nervous system, especially in relation to neurons, the negative effects of ethanol on the release of glutamate might affect neuron-glia communication in the hippocampus, which might lead to functional defects in the brain.
Collapse
|
32
|
Glutathione Modulates Ca2+ Influx and Oxidative Toxicity Through TRPM2 Channel in Rat Dorsal Root Ganglion Neurons. J Membr Biol 2011; 242:109-18. [DOI: 10.1007/s00232-011-9382-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 06/17/2011] [Indexed: 01/25/2023]
|
33
|
Tokuda K, Izumi Y, Zorumski CF. Ethanol enhances neurosteroidogenesis in hippocampal pyramidal neurons by paradoxical NMDA receptor activation. J Neurosci 2011; 31:9905-9. [PMID: 21734282 PMCID: PMC3180997 DOI: 10.1523/jneurosci.1660-11.2011] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 05/06/2011] [Accepted: 05/24/2011] [Indexed: 01/08/2023] Open
Abstract
Using an antibody against 5α-reduced neurosteroids, predominantly allopregnanolone, we found that immunostaining in the CA1 region of rat hippocampal slices was confined to pyramidal neurons. This neurosteroid staining was increased following 15 min administration of 60 mm but not 20 mm ethanol, and the enhancement was blocked by finasteride and dutasteride, selective inhibitors of 5α-reductase, a key enzyme required for allopregnanolone synthesis. Consistent with a prior report indicating that N-methyl-D-aspartate (NMDA) receptor (NMDAR) activation can promote steroid production, we observed that D-2-amino-5-phosphonovalerate (APV), a competitive NMDAR antagonist, blocked the effects of 60 mm ethanol on staining. We previously reported that 60 mm ethanol inhibits the induction of long-term potentiation (LTP), a cellular model for memory formation, in the CA1 region. In the present study, LTP inhibition by 60 mm ethanol was also overcome by both the 5α-reductase inhibitors and by APV. Furthermore, the effects of ethanol on neurosteroid production and LTP were mimicked by a low concentration of NMDA (1 μm), and the ability of NMDA to inhibit LTP and to enhance neurosteroid staining was reversed by finasteride and dutasteride, as well as by APV. These results indicate that ethanol paradoxically enhances GABAergic neurosteroid production by activation of unblocked NMDARs and that acute LTP inhibition by ethanol represents a form of NMDAR-mediated metaplasticity.
Collapse
Affiliation(s)
| | | | - Charles F. Zorumski
- Departments of Psychiatry and
- Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
34
|
Parkhomenko YM, Kudryavtsev PA, Pylypchuk SY, Chekhivska LI, Stepanenko SP, Sergiichuk AA, Bunik VI. Chronic alcoholism in rats induces a compensatory response, preserving brain thiamine diphosphate, but the brain 2-oxo acid dehydrogenases are inactivated despite unchanged coenzyme levels. J Neurochem 2011; 117:1055-65. [DOI: 10.1111/j.1471-4159.2011.07283.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Del Castillo-Vaquero A, Salido GM, González A. Increased calcium influx in the presence of ethanol in mouse pancreatic acinar cells. Int J Exp Pathol 2010; 91:114-124. [PMID: 20002836 PMCID: PMC2965897 DOI: 10.1111/j.1365-2613.2009.00691.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 09/15/2009] [Indexed: 12/15/2022] Open
Abstract
The effects of alcohol on Ca(2+) signalling remains poorly understood. Here we have investigated the effects of acute ethanol exposure on Ca(2+) influx in mouse pancreatic acinar cells. Cells were loaded with fura-2 and the changes in fluorescence were monitored by spectrofluorimetry and imaging analysis. Stimulation of cells with 20 pM cholecystokinin evoked an oscillatory pattern in [Ca(2+)](c), both in the presence and in the absence of extracellular Ca(2+). Stimulation of cells with cholecystokinin in the presence of 50 mM ethanol led to a transformation of physiological oscillations into a single transient increase in [Ca(2+)](c). This effect was observed when Ca(2+) was present in the extracellular medium, and did not appear in its absence. Addition of 1 mM CaCl(2) to the extracellular medium, following release of Ca(2+) from intracellular stores by stimulation of cells with 1 nM cholecystokinin or 1 microM thapsigargin in the absence of extracellular Ca(2+), was followed by an increase in [Ca(2+)](c). Ca(2+) influx was increased in the presence of 50 mM ethanol. The anti-oxidant cinnamtannin B-1 (10 microM) or inhibition of alcohol dehydrogenase by 4-MP (1 mM), significantly reduced Ca(2+) influx evoked by cholecystokinin in the presence of ethanol. In summary, intoxicating concentrations of ethanol may lead to over stimulation of pancreatic acinar cells by cholecystokinin. This might be partially explained by the generation of reactive oxygen species and an increased Ca(2+) entry in the presence of ethanol. Potentially ethanol might lead to Ca(2+) overload, which is a common pathological precursor that is implicated in pancreatitis.
Collapse
|
36
|
Yang H, Cui GB, Jiao XY, Wang J, Ju G, You SW. Thymosin-beta4 attenuates ethanol-induced neurotoxicity in cultured cerebral cortical astrocytes by inhibiting apoptosis. Cell Mol Neurobiol 2010; 30:149-60. [PMID: 19688260 PMCID: PMC11498471 DOI: 10.1007/s10571-009-9439-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Accepted: 07/27/2009] [Indexed: 12/26/2022]
Abstract
Thymosin-beta4 (Tbeta4) is a major actin monomer-binding peptide in mammalian tissues and plays a crucial role in the nervous system in synaptogenesis, neuronal survival and migration, axonal growth, and plastic changes of dendritic spines. However, it is unknown whether Tbeta4 is also involved in challenges with external stress such as ethanol-induced neurotoxicity. In the present study, we investigated the effects of Tbeta4 on ethanol-induced neurotoxicity in cultured cerebral cortical astrocytes and the underlying mechanisms. Primarily cultured astrocytes were treated with 1 microg/ml Tbeta4 2 h prior to administration of 100 mM ethanol for 0.5, 1, 3 and 6 days, respectively. The results showed that ethanol caused neurotoxicity in cultured astrocytes, as shown by declined cell viability, distinct astroglial apoptosis and increased intracellular peroxidation. Tbeta4 markedly promoted cell viability, ameliorated the injury of intracellular glial fibrillary acidic protein-immunopositive cytoskeletal structures, reduced the percentage of apoptotic astrocyte and cellular DNA fragmentation, suppressed caspase-3 activity and upregulated Bcl-2 expression, inhibited the accumulation of reactive oxygen species and production of malondialdehyde in ethanol-treated astrocytes in a time-dependent manner. These data indicated that Tbeta4 attenuates ethanol-induced neurotoxicity in cultured cortical astrocytes through inhibition of apoptosis signaling, and one of the mechanisms underlying the capacity of Tbeta4 to suppress apoptosis may in part be due to its effect of anti-peroxidation.
Collapse
Affiliation(s)
- Hao Yang
- Institute of Neurosciences, The Fourth Military Medical University, 710032 Xi’an, China
| | - Guang-Bin Cui
- Department of Radiology, Tangdu Hospital, The Fourth Military Medical University, 710038 Xi’an, China
| | - Xi-Ying Jiao
- Institute of Neurosciences, The Fourth Military Medical University, 710032 Xi’an, China
| | - Jian Wang
- Institute of Neurosciences, The Fourth Military Medical University, 710032 Xi’an, China
| | - Gong Ju
- Institute of Neurosciences, The Fourth Military Medical University, 710032 Xi’an, China
| | - Si-Wei You
- Institute of Neurosciences, The Fourth Military Medical University, 710032 Xi’an, China
| |
Collapse
|
37
|
MK801 blocks hypoxic blood–brain-barrier disruption and leukocyte adhesion. Neurosci Lett 2009; 449:168-72. [DOI: 10.1016/j.neulet.2008.10.096] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 10/20/2008] [Accepted: 10/21/2008] [Indexed: 11/24/2022]
|
38
|
González A, Salido GM. Ethanol alters the physiology of neuron-glia communication. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 88:167-98. [PMID: 19897078 DOI: 10.1016/s0074-7742(09)88007-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the central nervous system (CNS), both neurones and astrocytes play crucial roles. On a cellular level, brain activity involves continuous interactions within complex cellular circuits established between neural cells and glia. Although it was initially considered that neurones were the major cell type in cerebral function, nowadays astrocytes are considered to contribute to cerebral function too. Astrocytes support normal neuronal activity, including synaptic function, by regulating the extracellular environment with respect to ions and neurotransmitters. There is a plethora of noxious agents which can lead to the development of alterations in organs and functional systems, and that will end in a chronic prognosis. Among the potentially harmful external agents we can find ethanol consumption, whose consequences have been recognized as a major public health concern. Deregulation of cell cycle has devastating effects on the integrity of cells, and has been closely associated with the development of pathologies which can lead to dysfunction and cell death. An alteration of normal neuronal-glial physiology could represent the basis of neurodegenerative processes. In this review we will pay attention on to the recent findings in astrocyte function and their role toward neurons under ethanol consumption.
Collapse
Affiliation(s)
- Antonio González
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10071, Cáceres, Spain
| | | |
Collapse
|