1
|
Namba MD, Xie Q, Park K, Jackson JG, Barker JM. EcoHIV Infection Modulates the Effects of Cocaine Exposure Pattern and Abstinence on Cocaine Seeking and Neuroimmune Protein Expression in Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589615. [PMID: 38659915 PMCID: PMC11042347 DOI: 10.1101/2024.04.15.589615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cocaine use disorders (CUDs) and human immunodeficiency virus (HIV) remain persistent public health dilemmas throughout the world. One major hurdle for treating CUD is the increase in cocaine craving and seeking behavior that occurs over a protracted period of abstinence, an effect known as the incubation of craving. Little is known about how HIV may modulate this process. Thus, we sought to examine the impact of chronic HIV infection on the incubation of cocaine craving and associated changes in the central and peripheral immune systems. Here, mice were inoculated with EcoHIV, which is a chimeric HIV-1 construct that produces chronic HIV infection in mice. EcoHIV- and sham-infected mice were conditioned with cocaine daily or intermittently in a conditioned place preference (CPP) paradigm, followed by 1 or 21 days of forced abstinence prior to assessing preference for the cocaine-paired chamber. Under both conditioning regimens, sham mice exhibited incubation of cocaine CPP after 21 days of abstinence. EcoHIV-infected mice conditioned daily with cocaine showed enhanced cocaine seeking at both abstinence timepoints, whereas infected mice conditioned intermittently showed a reversal of the incubation effect, with higher cocaine seeking after 1 day of abstinence compared to 21 days. Analysis of corticolimbic CX3CL1-CX3CR1 and glutamate receptor expression revealed alterations in medial prefrontal cortex (mPFC) CX3CL1 and nucleus accumbens (NAc) GluN2A receptors that correlated with cocaine seeking following daily cocaine exposure. Moreover, examination of peripheral immune markers showed that the effect of abstinence and EcoHIV infection on these measures depended on the cocaine exposure regimen. Altogether, these results highlight the importance of cocaine abstinence and exposure pattern as critical variables that modulate HIV-associated neuroimmune outcomes and relapse vulnerability.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
- Graduate Program in Pharmacology and Physiology, College of Medicine, Drexel University
| | - Kyewon Park
- Center for AIDS Research (CFAR), University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua G. Jackson
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M. Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Gruol DL, Calderon D, Huitron-Resendiz S, Cates-Gatto C, Roberts AJ. Impact of Elevated Brain IL-6 in Transgenic Mice on the Behavioral and Neurochemical Consequences of Chronic Alcohol Exposure. Cells 2023; 12:2306. [PMID: 37759527 PMCID: PMC10527024 DOI: 10.3390/cells12182306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Alcohol consumption activates the neuroimmune system of the brain, a system in which brain astrocytes and microglia play dominant roles. These glial cells normally produce low levels of neuroimmune factors, which are important signaling factors and regulators of brain function. Alcohol activation of the neuroimmune system is known to dysregulate the production of neuroimmune factors, such as the cytokine IL-6, thereby changing the neuroimmune status of the brain, which could impact the actions of alcohol. The consequences of neuroimmune-alcohol interactions are not fully known. In the current studies we investigated this issue in transgenic (TG) mice with altered neuroimmune status relative to IL-6. The TG mice express elevated levels of astrocyte-produced IL-6, a condition known to occur with alcohol exposure. Standard behavioral tests of alcohol drinking and negative affect/emotionality were carried out in homozygous and heterozygous TG mice and control mice to assess the impact of neuroimmune status on the actions of chronic intermittent alcohol (ethanol) (CIE) exposure on these behaviors. The expressions of signal transduction and synaptic proteins were also assessed by Western blot to identify the impact of alcohol-neuroimmune interactions on brain neurochemistry. The results from these studies show that neuroimmune status with respect to IL-6 significantly impacts the effects of alcohol on multiple levels.
Collapse
Affiliation(s)
- Donna L. Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Delilah Calderon
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA 92037, USA (A.J.R.)
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA 92037, USA (A.J.R.)
| |
Collapse
|
3
|
Huffels CFM, Middeldorp J, Hol EM. Aß Pathology and Neuron-Glia Interactions: A Synaptocentric View. Neurochem Res 2023; 48:1026-1046. [PMID: 35976488 PMCID: PMC10030451 DOI: 10.1007/s11064-022-03699-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 10/15/2022]
Abstract
Alzheimer's disease (AD) causes the majority of dementia cases worldwide. Early pathological hallmarks include the accumulation of amyloid-ß (Aß) and activation of both astrocytes and microglia. Neurons form the building blocks of the central nervous system, and astrocytes and microglia provide essential input for its healthy functioning. Their function integrates at the level of the synapse, which is therefore sometimes referred to as the "quad-partite synapse". Increasing evidence puts AD forward as a disease of the synapse, where pre- and postsynaptic processes, as well as astrocyte and microglia functioning progressively deteriorate. Here, we aim to review the current knowledge on how Aß accumulation functionally affects the individual components of the quad-partite synapse. We highlight a selection of processes that are essential to the healthy functioning of the neuronal synapse, including presynaptic neurotransmitter release and postsynaptic receptor functioning. We further discuss how Aß affects the astrocyte's capacity to recycle neurotransmitters, release gliotransmitters, and maintain ion homeostasis. We additionally review literature on how Aß changes the immunoprotective function of microglia during AD progression and conclude by summarizing our main findings and highlighting the challenges in current studies, as well as the need for further research.
Collapse
Affiliation(s)
- Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
4
|
Bellettini-Santos T, Garcez ML, Mina F, Magnus NQ, Pereira NDS, Marques ADO, Keller GS, Zabot GC, do Nascimento NB, Medeiros EB, Rempel LCT, Kucharska E, Frizon TEA, Dal-Bó AG, Budni J. Vitamin D3 improves spatial memory and modulates cytokine levels in aged rats. Metab Brain Dis 2023; 38:1155-1166. [PMID: 36689104 DOI: 10.1007/s11011-022-01152-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/21/2022] [Indexed: 01/24/2023]
Abstract
Vitamin D3 deficiency is associated with an increased risk of dementia. An association between vitamin D3 deficiency and subjective cognitive complaints in geriatric patients has been previously reported. This study aimed to evaluate the effects of two doses of vitamin D3 on spatial memory (using the Radial Maze) and cytokine levels [tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-10 (IL-10)] on 2-, 6-, 13-, 22-, and 31-month-old male Wistar rats. Animals were supplemented with vitamin D3 at doses of 42 IU/kg and 420 IU/kg for 21 days. A radial maze test was performed to evaluate spatial memory. After the behavioral test, the frontal cortex and hippocampus were dissected for enzyme immunoassay analyses to measure the cytokine levels (TNFα, IL-1β, IL-6, and IL-10). Our results showed that vitamin D3 supplementation reversed spatial memory impairment at the supplemented doses (42 and 420 IU/kg) in 6-, 13-, and 22-month-old animals and at a dose of 420 IU/kg in 31-month-old animals. The lower dose (42 IU/kg) regulates both pro- and anti-inflammatory cytokines mainly in the frontal cortex. Our results suggest that vitamin D3 has a modulatory action on pro- and anti-inflammatory cytokines, since older animals showed increased cytokine levels compared to 2-month-old animals, and that vitamin D3 may exert an immunomodulatory effect on aging.
Collapse
Affiliation(s)
- Tatiani Bellettini-Santos
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, ES, Brazil
| | - Michelle Lima Garcez
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Colatina, ES, Brazil
| | - Francielle Mina
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Natália Quadros Magnus
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Nathalia de Souza Pereira
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Ariandne de Oliveira Marques
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Gabriela Serafim Keller
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Gabriel Casagrande Zabot
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Natália Baltazar do Nascimento
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Eduarda Behenck Medeiros
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Lisienny Campoli Tono Rempel
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil
| | - Ewa Kucharska
- Department of Health Education, Gerontology and Geriatrics, Jesuit University Ignatianum in Krakow, 31-501, Krakow, Poland
| | - Tiago Elias Allievi Frizon
- Laboratory of Advanced Polymer Processing, Universidade Do Extremo Sul Catarinense - UNESC, Criciúma, SC, Brazil
- Department of Energy and Sustainability, Federal University of Santa Catarina - UFSC, Araranguá, SC, Brazil
| | - Alexandre Gonçalves Dal-Bó
- Laboratory of Advanced Polymer Processing, Universidade Do Extremo Sul Catarinense - UNESC, Criciúma, SC, Brazil
| | - Josiane Budni
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
5
|
Repositioning doxycycline for treating synucleinopathies: Evidence from a pre-clinical mouse model. Parkinsonism Relat Disord 2023; 106:105229. [PMID: 36462409 DOI: 10.1016/j.parkreldis.2022.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Parkinson's disease remains orphan of valuable therapies capable to interfere with the disease pathogenesis despite the large number of symptomatic approaches adopted in clinical practice to manage this disease. Treatments simultaneously affecting α-synuclein (α-syn) oligomerization and neuroinflammation may counteract Parkinson's disease and related disorders. Recent data demonstrate that Doxycycline, a tetracycline antibiotic, can inhibit α-syn aggregation as well as neuroinflammation. We herein investigate, for the first time, the potential therapeutic properties of Doxy in a human α-syn A53T transgenic Parkinson's disease mouse model evaluating behavioural, biochemical and histopathological parameters. EXPERIMENTAL APPROACH Human α-syn A53T transgenic mice were treated with Doxycycline (10 mg/kg daily ip) for 30 days. The effect of treatment on motor, cognitive and daily live activity performances were examined. Neuropathological and neurophysiological parameters were assessed through immunocytochemical, electrophysiological and biochemical analysis of cerebral tissue. KEY RESULTS Doxy treatment abolished cognitive and daily life activity deficiencies in A53T mice. The effect on cognitive functions was associated with neuroprotection, inhibition of α-syn oligomerization and gliosis both in the cortex and hippocampus. Doxy treatment restored hippocampal long-term potentiation in association with the inhibition of pro-inflammatory cytokines expression. Moreover, Doxy ameliorated motor impairment and reduced striatal glial activation in A53T mice. CONCLUSIONS AND IMPLICATIONS Our findings promote Doxy as a valuable multi-target therapeutic approach counteracting both symptoms and neuropathology in the complex scenario of α-synucleinopathies.
Collapse
|
6
|
De Felice E, Gonçalves de Andrade E, Golia MT, González Ibáñez F, Khakpour M, Di Castro MA, Garofalo S, Di Pietro E, Benatti C, Brunello N, Tascedda F, Kaminska B, Limatola C, Ragozzino D, Tremblay ME, Alboni S, Maggi L. Microglial diversity along the hippocampal longitudinal axis impacts synaptic plasticity in adult male mice under homeostatic conditions. J Neuroinflammation 2022; 19:292. [PMID: 36482444 PMCID: PMC9730634 DOI: 10.1186/s12974-022-02655-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.
Collapse
Affiliation(s)
- E. De Felice
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Gonçalves de Andrade
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. T. Golia
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - F. González Ibáñez
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - M. Khakpour
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. A. Di Castro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - S. Garofalo
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Di Pietro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - C. Benatti
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N. Brunello
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F. Tascedda
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - B. Kaminska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - C. Limatola
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - D. Ragozzino
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy ,grid.417778.a0000 0001 0692 3437Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - M. E. Tremblay
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - S. Alboni
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - L. Maggi
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
7
|
Hikosaka M, Kawano T, Wada Y, Maeda T, Sakurai T, Ohtsuki G. Immune-Triggered Forms of Plasticity Across Brain Regions. Front Cell Neurosci 2022; 16:925493. [PMID: 35978857 PMCID: PMC9376917 DOI: 10.3389/fncel.2022.925493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/16/2022] [Indexed: 01/03/2023] Open
Abstract
Immune cells play numerous roles in the host defense against the invasion of microorganisms and pathogens, which induces the release of inflammatory mediators (e.g., cytokines and chemokines). In the CNS, microglia is the major resident immune cell. Recent efforts have revealed the diversity of the cell types and the heterogeneity of their functions. The refinement of the synapse structure was a hallmark feature of the microglia, while they are also involved in the myelination and capillary dynamics. Another promising feature is the modulation of the synaptic transmission as synaptic plasticity and the intrinsic excitability of neurons as non-synaptic plasticity. Those modulations of physiological properties of neurons are considered induced by both transient and chronic exposures to inflammatory mediators, which cause behavioral disorders seen in mental illness. It is plausible for astrocytes and pericytes other than microglia and macrophage to induce the immune-triggered plasticity of neurons. However, current understanding has yet achieved to unveil what inflammatory mediators from what immune cells or glia induce a form of plasticity modulating pre-, post-synaptic functions and intrinsic excitability of neurons. It is still unclear what ion channels and intracellular signaling of what types of neurons in which brain regions of the CNS are involved. In this review, we introduce the ubiquitous modulation of the synaptic efficacy and the intrinsic excitability across the brain by immune cells and related inflammatory cytokines with the mechanism for induction. Specifically, we compare neuro-modulation mechanisms by microglia of the intrinsic excitability of cerebellar Purkinje neurons with cerebral pyramidal neurons, stressing the inverted directionality of the plasticity. We also discuss the suppression and augmentation of the extent of plasticity by inflammatory mediators, as the meta-plasticity by immunity. Lastly, we sum up forms of immune-triggered plasticity in the different brain regions with disease relevance. Together, brain immunity influences our cognition, sense, memory, and behavior via immune-triggered plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | - Gen Ohtsuki
- Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
8
|
Mezo-González CE, Daher Abdi A, Reyes-Castro LA, Olvera Hernández S, Almeida C, Croyal M, Aguesse A, Gavioli EC, Zambrano E, Bolaños-Jiménez F. Learning Deficits Induced by High-Calorie Feeding in the Rat are Associated With Impaired Brain Kynurenine Pathway Metabolism. Int J Tryptophan Res 2022; 15:11786469221111116. [PMID: 35846874 PMCID: PMC9277427 DOI: 10.1177/11786469221111116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
In addition to be a primary risk factor for type 2 diabetes and cardiovascular
disease, obesity is associated with learning disabilities. Here we examined
whether a dysregulation of the kynurenine pathway (KP) of tryptophan (Trp)
metabolism might underlie the learning deficits exhibited by obese individuals.
The KP is initiated by the enzymatic conversion of Trp into kynurenine (KYN) by
indoleamine 2,3-dioxygenase (IDO). KYN is further converted to several signaling
molecules including quinolinic acid (QA) which has a negative impact on
learning. Wistar rats were fed either standard chow or made obese by exposure to
a free choice high-fat high-sugar (fcHFHS) diet. Their learning capacities were
evaluated using a combination of the novel object recognition and the novel
object location tasks, and the concentrations of Trp and KYN-derived metabolites
in several brain regions determined by ultra-performance liquid
chromatography-tandem mass spectrometry. Male, but not female, obese rats
exhibited reduced learning capacity characterized by impaired encoding along
with increased hippocampal concentrations of QA, Xanthurenic acid (XA),
Nicotinamide (Nam), and oxidized Nicotinamide Adenine Dinucleotide (NAD+). In
contrast, no differences were detected in the serum levels of Trp or KP
metabolites. Moreover, obesity enhanced the expression in the hippocampus and
frontal cortex of kynurenine monooxygenase (KMO), an enzyme involved in the
production of QA from kynurenine. QA stimulates the glutamatergic system and its
increased production leads to cognitive impairment. These results suggest that
the deleterious effects of obesity on cognition are sex dependent and that
altered KP metabolism might contribute to obesity-associated learning
disabilities.
Collapse
Affiliation(s)
| | - Amran Daher Abdi
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Université de Nantes, Nantes France
| | - Luis Antonio Reyes-Castro
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Université de Nantes, Nantes France.,Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Sandra Olvera Hernández
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Université de Nantes, Nantes France.,Medical and Psychology School, Autonomous University of Baja California, Tijuana, Mexico
| | - Clarissa Almeida
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Mikaël Croyal
- CRNH-O Mass Spectrometry Core Facility, Nantes, France.,Université de Nantes, CNRS, INSERM, L'institut du Thorax, Nantes, France.,Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | | | - Elaine Cristina Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | |
Collapse
|
9
|
Lyra E Silva NM, Barros-Aragão FGQ, De Felice FG, Ferreira ST. Inflammation at the crossroads of COVID-19, cognitive deficits and depression. Neuropharmacology 2022; 209:109023. [PMID: 35257690 PMCID: PMC8894741 DOI: 10.1016/j.neuropharm.2022.109023] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Natalia M Lyra E Silva
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada.
| | - Fernanda G Q Barros-Aragão
- D'OR Institute for Research & Education, RJ, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil.
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada; D'OR Institute for Research & Education, RJ, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil
| | - Sergio T Ferreira
- D'OR Institute for Research & Education, RJ, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ, Brazil
| |
Collapse
|
10
|
Sanguino‐Gómez J, Buurstede JC, Abiega O, Fitzsimons CP, Lucassen PJ, Eggen BJL, Lesuis SL, Meijer OC, Krugers HJ. An emerging role for microglia in stress‐effects on memory. Eur J Neurosci 2021; 55:2491-2518. [PMID: 33724565 PMCID: PMC9373920 DOI: 10.1111/ejn.15188] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/13/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Stressful experiences evoke, among others, a rapid increase in brain (nor)epinephrine (NE) levels and a slower increase in glucocorticoid hormones (GCs) in the brain. Microglia are key regulators of neuronal function and contain receptors for NE and GCs. These brain cells may therefore potentially be involved in modulating stress effects on neuronal function and learning and memory. In this review, we discuss that stress induces (1) an increase in microglial numbers as well as (2) a shift toward a pro‐inflammatory profile. These microglia have (3) impaired crosstalk with neurons and (4) disrupted glutamate signaling. Moreover, microglial immune responses after stress (5) alter the kynurenine pathway through metabolites that impair glutamatergic transmission. All these effects could be involved in the impairments in memory and in synaptic plasticity caused by (prolonged) stress, implicating microglia as a potential novel target in stress‐related memory impairments.
Collapse
Affiliation(s)
| | - Jacobus C. Buurstede
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Oihane Abiega
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems Section Molecular Neurobiology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
- Program in Neurosciences and Mental Health Hospital for Sick Children Toronto ON Canada
| | - Onno C. Meijer
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
11
|
Namba MD, Leyrer-Jackson JM, Nagy EK, Olive MF, Neisewander JL. Neuroimmune Mechanisms as Novel Treatment Targets for Substance Use Disorders and Associated Comorbidities. Front Neurosci 2021; 15:650785. [PMID: 33935636 PMCID: PMC8082184 DOI: 10.3389/fnins.2021.650785] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies examining the neurobiology of substance abuse have revealed a significant role of neuroimmune signaling as a mechanism through which drugs of abuse induce aberrant changes in synaptic plasticity and contribute to substance abuse-related behaviors. Immune signaling within the brain and the periphery critically regulates homeostasis of the nervous system. Perturbations in immune signaling can induce neuroinflammation or immunosuppression, which dysregulate nervous system function including neural processes associated with substance use disorders (SUDs). In this review, we discuss the literature that demonstrates a role of neuroimmune signaling in regulating learning, memory, and synaptic plasticity, emphasizing specific cytokine signaling within the central nervous system. We then highlight recent preclinical studies, within the last 5 years when possible, that have identified immune mechanisms within the brain and the periphery associated with addiction-related behaviors. Findings thus far underscore the need for future investigations into the clinical potential of immunopharmacology as a novel approach toward treating SUDs. Considering the high prevalence rate of comorbidities among those with SUDs, we also discuss neuroimmune mechanisms of common comorbidities associated with SUDs and highlight potentially novel treatment targets for these comorbid conditions. We argue that immunopharmacology represents a novel frontier in the development of new pharmacotherapies that promote long-term abstinence from drug use and minimize the detrimental impact of SUD comorbidities on patient health and treatment outcomes.
Collapse
Affiliation(s)
- Mark D. Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Erin K. Nagy
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | | |
Collapse
|
12
|
Wegrzyn D, Freund N, Faissner A, Juckel G. Poly I:C Activated Microglia Disrupt Perineuronal Nets and Modulate Synaptic Balance in Primary Hippocampal Neurons in vitro. Front Synaptic Neurosci 2021; 13:637549. [PMID: 33708102 PMCID: PMC7940526 DOI: 10.3389/fnsyn.2021.637549] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/03/2021] [Indexed: 12/31/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized, reticular structures of the extracellular matrix (ECM) that can be found covering the soma and proximal dendrites of a neuronal subpopulation. Recent studies have shown that PNNs can highly influence synaptic plasticity and are disrupted in different neuropsychiatric disorders like schizophrenia. Interestingly, there is a growing evidence that microglia can promote the loss of PNNs and contribute to neuropsychiatric disorders. Based on this knowledge, we analyzed the impact of activated microglia on hippocampal neuronal networks in vitro. Therefore, primary cortical microglia were cultured and stimulated via polyinosinic-polycytidylic acid (Poly I:C; 50 μg/ml) administration. The Poly I:C treatment induced the expression and secretion of different cytokines belonging to the CCL- and CXCL-motif chemokine family as well as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). In addition, the expression of matrix metalloproteinases (MMPs) could be verified via RT-PCR analysis. Embryonic hippocampal neurons were then cultured for 12 days in vitro (DIV) and treated for 24 h with microglial conditioned medium. Interestingly, immunocytochemical staining of the PNN component Aggrecan revealed a clear disruption of PNNs accompanied by a significant increase of glutamatergic and a decrease of γ-aminobutyric acid-(GABA)ergic synapse numbers on PNN wearing neurons. In contrast, PNN negative neurons showed a significant reduction in both, glutamatergic and GABAergic synapses. Electrophysiological recordings were performed via multielectrode array (MEA) technology and unraveled a significantly increased spontaneous network activity that sustained also 24 and 48 h after the administration of microglia conditioned medium. Taken together, we could observe a strong impact of microglial secreted factors on PNN integrity, synaptic plasticity and electrophysiological properties of cultured neurons. Our observations might enhance the understanding of neuron-microglia interactions considering the ECM.
Collapse
Affiliation(s)
- David Wegrzyn
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Georg Juckel
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
13
|
La Vitola P, Balducci C, Baroni M, Artioli L, Santamaria G, Castiglioni M, Cerovic M, Colombo L, Caldinelli L, Pollegioni L, Forloni G. Peripheral inflammation exacerbates α-synuclein toxicity and neuropathology in Parkinson's models. Neuropathol Appl Neurobiol 2021; 47:43-60. [PMID: 32696999 DOI: 10.1111/nan.12644] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
AIMS Parkinson's disease and related disorders are devastating neurodegenerative pathologies. Since α-synuclein was identified as a main component of Lewy bodies and neurites, efforts have been made to clarify the pathogenic mechanisms of α-synuclein's detrimental effects. α-synuclein oligomers are the most harmful species and may recruit and activate glial cells. Inflammation is emerging as a bridge between genetic susceptibility and environmental factors co-fostering Parkinson's disease. However, direct evidence linking inflammation to the harmful activities of α-synuclein oligomers or to the Parkinson's disease behavioural phenotype is lacking. METHODS To clarify whether neuroinflammation influences Parkinson's disease pathogenesis, we developed: (i) a 'double-hit' approach in C57BL/6 naive mice where peripherally administered lipopolysaccharides were followed by intracerebroventricular injection of an inactive oligomer dose; (ii) a transgenic 'double-hit' model where lipopolysaccharides were given to A53T α-synuclein transgenic Parkinson's disease mice. RESULTS Lipopolysaccharides induced a long-lasting neuroinflammatory response which facilitated the detrimental cognitive activities of oligomers. LPS-activated microglia and astrocytes responded differently to the oligomers with microglia activating further and acquiring a pro-inflammatory M1 phenotype, while astrocytes atrophied. In the transgenic 'double-hit' A53T mouse model, lipopolysaccharides aggravated cognitive deficits and increased microgliosis. Again, astrocytes responded differently to the double challenge. These findings indicate that peripherally induced neuroinflammation potentiates the α-synuclein oligomer's actions and aggravates cognitive deficits in A53T mice. CONCLUSIONS The fine management of both peripheral and central inflammation may offer a promising therapeutic approach to prevent or slow down some behavioural aspects in α-synucleinopathies.
Collapse
Affiliation(s)
- P La Vitola
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - C Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Baroni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Artioli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - G Santamaria
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Castiglioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Cerovic
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Caldinelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - L Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - G Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
14
|
Loprinzi PD, Moore D, Loenneke JP. Does Aerobic and Resistance Exercise Influence Episodic Memory through Unique Mechanisms? Brain Sci 2020; 10:E913. [PMID: 33260817 PMCID: PMC7761124 DOI: 10.3390/brainsci10120913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Aerobic and resistance exercise (acute and chronic) independently and collectively induce beneficial responses in the brain that may influence memory function, including an increase in cerebral blood flow, neurogenesis, neuroelectrical alterations, and protein production. However, whether aerobic and resistance exercise improve memory via similar or distinct mechanisms has yet to be fully explained. Here, we review the unique influence of aerobic and resistance exercise on neural modulation, proteins, receptors, and ultimately, episodic memory. Resistance training may optimize neural communication, information processing and memory encoding by affecting the allocation of attentional resources. Moreover, resistance exercise can reduce inflammatory markers associated with neural communication while increasing peripheral and central BDNF (brain-derived neurotrophic factor) production. Aerobic training increases hippocampal levels of BDNF and TrkB (Tropomyosin receptor kinase B), protein kinases and glutamatergic proteins. Likewise, both aerobic and anaerobic exercise can increase CREB (cAMP response element-binding protein) phosphorylation. Thus, we suggest that aerobic and resistance exercise may influence episodic memory via similar and, potentially, distinct mechanisms.
Collapse
Affiliation(s)
- Paul D. Loprinzi
- Exercise & Memory Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, Oxford, MS 38677, USA;
| | - Damien Moore
- Exercise & Memory Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, Oxford, MS 38677, USA;
| | - Jeremy P. Loenneke
- Kevser Ermin Applied Physiology Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
15
|
Interleukin-6: A neuro-active cytokine contributing to cognitive impairment in Duchenne muscular dystrophy? Cytokine 2020; 133:155134. [DOI: 10.1016/j.cyto.2020.155134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
|
16
|
Divanbeigi A, Nasehi M, Vaseghi S, Amiri S, Zarrindast MR. Tropisetron But Not Granisetron Ameliorates Spatial Memory Impairment Induced by Chronic Cerebral Hypoperfusion. Neurochem Res 2020; 45:2631-2640. [PMID: 32797381 DOI: 10.1007/s11064-020-03110-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/16/2022]
Abstract
Tropisetron and Granisetorn are 5-HT3 antagonists with antiemetic effects. Tropisetron also has a partial agonistic effect on alpha-7 nicotinic acetylcholine receptors (α7 nAChRs). On the other hand, chronic cerebral hypoperfusion (CCH) attenuates cerebral blood flow and impairs cognitive functions. The goal of this study was to investigate the effect of Tropisetron and Granisetron on CCH-induced spatial memory impairment in rats. Forty-eight male Wistar rats were used in this study. 2-VO surgery was done to induce CCH and Radial Eight Arm Maz apparatus was used to evaluate spatial memory (working and reference memory). Tropisetron was injected intraperitoneally at the doses of 1 and 5 mg/kg, and Granisetron was injected intraperitoneally at the dose of 3 mg/kg. Dorsal hippocampal (CA1) neurons count, Interleukin 6 (IL-6) serum level, and serotonin-reuptake transporter (SERT) gene expression were also evaluated. The results showed, CCH impaired working and reference memory, increased IL-6 serum level, and decreased CA1 neurons and SERT expression. Tropisetron at the dose of 5 mg/kg restored all the effects of CCH. However, Granisetron did not restore CCH-induced memory impairment. Furthermore, Granisetron had no effect on IL-6. While, it increased SERT expression and CA1 neurons. In conclusion, Tropisetron but not Granisetron, ameliorated spatial memory impairment induced by CCH. We suggested conducting more detailed studies investigating the role of serotonergic system (5-HT3 receptors and serotonin transporters) and also α7 nAChRs in the effects of Tropisetron.
Collapse
Affiliation(s)
- Ashkan Divanbeigi
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran.,Scientific Research Committee, Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, P.O. Box 13145-784, Tehran, Iran.
| | - Salar Vaseghi
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran.,Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, P.O. Box 13145-784, Tehran, Iran
| | - Sepideh Amiri
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran.,Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, P.O. Box 13145-784, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Gruol DL, Hernandez RV, Roberts A. Alcohol Enhances Responses to High Frequency Stimulation in Hippocampus from Transgenic Mice with Increased Astrocyte Expression of IL-6. Cell Mol Neurobiol 2020; 41:1299-1310. [PMID: 32562098 DOI: 10.1007/s10571-020-00902-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Recent studies show that alcohol exposure can induce glial production of neuroimmune factors in the CNS. Of these, IL-6 has gained attention because it is involved in a number of important physiological and pathophysiological processes that could be affected by alcohol-induced CNS production of IL-6, particularly under conditions of excessive alcohol use. For example, IL-6 has been shown to play a role in hippocampal behaviors and synaptic plasticity (long-term potentiation; LTP) associated with memory and learning. Surprisingly, in our in vitro studies of LTP at the Schaffer collateral to CA1 pyramidal neuron synapse in hippocampus from transgenic mice that express elevated levels of astrocyte produced IL-6 (TG), LTP was not altered by the increased levels of IL-6. However, exposure to acute alcohol revealed neuroadaptive changes that served to protect LTP against the alcohol-induced reduction of LTP observed in hippocampus from non-transgenic control mice (WT). Here we examined the induction phase of LTP to assess if presynaptic neuroadaptive changes occurred in the hippocampus of TG mice that contributed to the resistance of LTP to alcohol. Results are consistent with a role for IL-6-induced neuroadaptive effects on presynaptic mechanisms involved in transmitter release in the resistance of LTP to alcohol in hippocampus from the TG mice. These actions are important with respect to a role for IL-6 in physiological and pathophysiological processes in the CNS and in CNS actions of alcohol, especially when excessive alcohol used is comorbid with conditions associated with elevated levels of IL-6 in the CNS.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Ruben V Hernandez
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
18
|
Devasahayam AJ, Chaves AR, Lasisi WO, Curtis ME, Wadden KP, Kelly LP, Pretty R, Chen A, Wallack EM, Newell CJ, Williams JB, Kenny H, Downer MB, McCarthy J, Moore CS, Ploughman M. Vigorous cool room treadmill training to improve walking ability in people with multiple sclerosis who use ambulatory assistive devices: a feasibility study. BMC Neurol 2020; 20:33. [PMID: 31969132 PMCID: PMC6975092 DOI: 10.1186/s12883-020-1611-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/10/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Aerobic training has the potential to restore function, stimulate brain repair, and reduce inflammation in people with Multiple Sclerosis (MS). However, disability, fatigue, and heat sensitivity are major barriers to exercise for people with MS. We aimed to determine the feasibility of conducting vigorous harness-supported treadmill training in a room cooled to 16 °C (10 weeks; 3times/week) and examine the longer-term effects on markers of function, brain repair, and inflammation among those using ambulatory aids. METHODS Ten participants (9 females) aged 29 to 74 years with an Expanded Disability Status Scale ranging from 6 to 7 underwent training (40 to 65% heart rate reserve) starting at 80% self-selected walking speed. Feasibility of conducting vigorous training was assessed using a checklist, which included attendance rates, number of missed appointments, reasons for not attending, adverse events, safety hazards during training, reasons for dropout, tolerance to training load, subjective reporting of symptom worsening during and after exercise, and physiological responses to exercise. Functional outcomes were assessed before, after, and 3 months after training. Walking ability was measured using Timed 25 Foot Walk test and on an instrumented walkway at both fast and self-selected speeds. Fatigue was measured using fatigue/energy/vitality sub-scale of 36-Item Short-Form (SF-36) Health Survey, Fatigue Severity Scale, modified Fatigue Impact Scale. Aerobic fitness (maximal oxygen consumption) was measured using maximal graded exercise test (GXT). Quality-of-life was measured using SF-36 Health Survey. Serum levels of neurotrophin (brain-derived neurotrophic factor) and cytokine (interleukin-6) were assessed before and after GXT. RESULTS Eight of the ten participants completed training (attendance rates ≥ 80%). No adverse events were observed. Fast walking speed (cm/s), gait quality (double-support (%)) while walking at self-selected speed, fatigue (modified Fatigue Impact Scale), fitness (maximal workload achieved during GXT), and quality-of-life (physical functioning sub-scale of SF-36) improved significantly after training, and improvements were sustained after 3-months. Improvements in fitness (maximal respiratory exchange ratio and maximal oxygen consumption during GXT) were associated with increased brain-derived neurotrophic factor and decreased interleukin-6. CONCLUSION Vigorous cool room training is feasible and can potentially improve walking, fatigue, fitness, and quality-of-life among people with moderate to severe MS-related disability. TRIAL REGISTRATION The study was approved by the Newfoundland and Labrador Health Research Ethics Board (reference number: 2018.088) on 11/07/2018 prior to the enrollment of first participant (retrospectively registered at ClinicalTrials.gov: NCT04066972. Registered on 26 August 2019.
Collapse
Affiliation(s)
- Augustine J Devasahayam
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Arthur R Chaves
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Wendy O Lasisi
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Marie E Curtis
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Katie P Wadden
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Liam P Kelly
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Ryan Pretty
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Alice Chen
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Elizabeth M Wallack
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Caitlin J Newell
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - John B Williams
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Rm H4360, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Hannah Kenny
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Matthew B Downer
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Jason McCarthy
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Rm H4360, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Michelle Ploughman
- Recovery & Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland, Rm 400, L.A. Miller Centre, 100 Forest Road, St. John's, NL, A1A 1E5, Canada.
| |
Collapse
|
19
|
Modeling Resilience to Damage in Multiple Sclerosis: Plasticity Meets Connectivity. Int J Mol Sci 2019; 21:ijms21010143. [PMID: 31878257 PMCID: PMC6981966 DOI: 10.3390/ijms21010143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/05/2019] [Accepted: 12/20/2019] [Indexed: 02/03/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by demyelinating white matter lesions and neurodegeneration, with a variable clinical course. Brain network architecture provides efficient information processing and resilience to damage. The peculiar organization characterized by a low number of highly connected nodes (hubs) confers high resistance to random damage. Anti-homeostatic synaptic plasticity, in particular long-term potentiation (LTP), represents one of the main physiological mechanisms underlying clinical recovery after brain damage. Different types of synaptic plasticity, including both anti-homeostatic and homeostatic mechanisms (synaptic scaling), contribute to shape brain networks. In MS, altered synaptic functioning induced by inflammatory mediators may represent a further cause of brain network collapse in addition to demyelination and grey matter atrophy. We propose that impaired LTP expression and pathologically enhanced upscaling may contribute to disrupting brain network topology in MS, weakening resilience to damage and negatively influencing the disease course.
Collapse
|
20
|
Tumor Necrosis Factor-α-Mediated Metaplastic Inhibition of LTP Is Constitutively Engaged in an Alzheimer's Disease Model. J Neurosci 2019; 39:9083-9097. [PMID: 31570539 DOI: 10.1523/jneurosci.1492-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
Abstract
LTP, a fundamental mechanism of learning and memory, is a highly regulated process. One form of regulation is metaplasticity (i.e., the activity-dependent and long-lasting changes in neuronal state that orchestrate the direction, magnitude, and persistence of future synaptic plasticity). We have previously described a heterodendritic metaplasticity effect, whereby strong high-frequency priming stimulation in stratum oriens inhibits subsequent LTP in the stratum radiatum of hippocampal area CA1, potentially by engagement of the enmeshed astrocytic network. This effect may occur due to neuron-glia interactions in response to priming stimulation that leads to the release of gliotransmitters. Here we found in male rats that TNFα and associated signal transduction enzymes, but not interleukin-1β (IL-1β), were responsible for mediating the metaplasticity effect. Replacing priming stimulation with TNFα incubation reproduced these effects. As TNFα levels are elevated in Alzheimer's disease, we examined whether heterodendritic metaplasticity is dysregulated in a transgenic mouse model of the disease, either before or after amyloid plaque formation. We showed that TNFα and IL-1β levels were significantly increased in aged but not young transgenic mice. Although control LTP was impaired in the young transgenic mice, it was not TNFα-dependent. In the older transgenic mice, however, LTP was impaired in a way that occluded further reduction by heterosynaptic metaplasticity, whereas LTP was entirely rescued by incubation with a TNFα antibody, but not an IL-1β antibody. Thus, TNFα mediates a heterodendritic metaplasticity in healthy rodents that becomes constitutively and selectively engaged in a mouse model of Alzheimer's disease.SIGNIFICANCE STATEMENT The proinflammatory cytokine TNFα is known to be capable of inhibiting LTP and is upregulated several-fold in brain tissue, serum, and CSF of Alzheimer's disease (AD) patients. However, the mechanistic roles played by TNFα in plasticity and AD remain poorly understood. Here we show that TNFα and its downstream signaling molecules p38 MAPK, ERK, and JNK contribute fundamentally to a long-range metaplastic inhibition of LTP in rats. Moreover, the impaired LTP in aged APP/PS1 mice is rescued by incubation with a TNFα antibody. Thus, there is an endogenous engagement of the metaplasticity mechanism in this mouse model of AD, supporting the idea that blocking TNFα might be of therapeutic benefit in the disease.
Collapse
|
21
|
Stampanoni Bassi M, Iezzi E, Mori F, Simonelli I, Gilio L, Buttari F, Sica F, De Paolis N, Mandolesi G, Musella A, De Vito F, Dolcetti E, Bruno A, Furlan R, Finardi A, Marfia GA, Centonze D, Rizzo FR. Interleukin-6 Disrupts Synaptic Plasticity and Impairs Tissue Damage Compensation in Multiple Sclerosis. Neurorehabil Neural Repair 2019; 33:825-835. [DOI: 10.1177/1545968319868713] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Synaptic plasticity helps in reducing the clinical expression of brain damage and represents a useful mechanism to compensate the negative impact of new brain lesions in multiple sclerosis (MS). Inflammation, altering synaptic plasticity, could negatively influence the disease course in relapsing-remitting MS (RR-MS). Objective: In the present study, we explored whether interleukin (IL)-6, a major proinflammatory cytokine involved in MS pathogenesis, alters synaptic plasticity and affects the ability to compensate for ongoing brain damage. Methods: The effect of IL-6 incubation on long-term potentiation (LTP) induction was explored in vitro, in mice hippocampal slices. We also explored the correlation between the cerebrospinal fluid (CSF) levels of this cytokine and the LTP-like effect induced by the paired associative stimulation (PAS) in a group of RR-MS patients. Finally, we examined the correlation between the CSF levels of IL-6 at the time of diagnosis and the prospective disease activity in a cohort of 150 RR-MS patients. Results: In vitro LTP induction was abolished by IL-6. Consistently, in patients with MS, a negative correlation emerged between IL-6 CSF concentrations and the effect of PAS. In MS patients, longer disease duration before diagnosis was associated with higher IL-6 CSF concentrations. In addition, elevated CSF levels of IL-6 were associated with greater clinical expression of new inflammatory brain lesions, unlike in patients with low or absent IL-6 concentrations, who had a better disease course. Conclusions: IL-6 interfering with synaptic plasticity mechanisms may impair the ability to compensate the clinical manifestation of new brain lesions in RR-MS patients.
Collapse
Affiliation(s)
- Mario Stampanoni Bassi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesco Mori
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | - Ilaria Simonelli
- Fondazione Fatebenefratelli per la Ricerca e la Formazione Sanitaria e Sociale, Rome, Italy
| | - Luana Gilio
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesco Sica
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Nicla De Paolis
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Georgia Mandolesi
- Laboratory of Synaptic Immunopathology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, Rome, Italy
- San Raffaele University, Via di Val Cannuta 247, Rome, Italy
| | - Alessandra Musella
- Laboratory of Synaptic Immunopathology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, Rome, Italy
- San Raffaele University, Via di Val Cannuta 247, Rome, Italy
| | - Francesca De Vito
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Ettore Dolcetti
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | - Antonio Bruno
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | | | | | - Girolama A. Marfia
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | - Diego Centonze
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli (IS), Italy
- Tor Vergata University, Department of Systems Medicine, Via Montpellier 1, Rome, Italy
| | | |
Collapse
|
22
|
Nenov MN, Malkov AE, Konakov MV, Levin SG. Interleukin-10 and transforming growth factor-β1 facilitate long-term potentiation in CA1 region of hippocampus. Biochem Biophys Res Commun 2019; 518:486-491. [PMID: 31434608 DOI: 10.1016/j.bbrc.2019.08.072] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022]
Abstract
It has been shown that pro-inflammatory cytokines preferentially attenuate long-term potentiation (LTP), at the same time the effect of anti-inflammatory cytokines on synaptic plasticity has not been fully studied yet. Here we studied the effect of two anti-inflammatory cytokines - interleukin-10 (IL-10) and transforming growth factor-β1 (TGF-β1) on long-term potentiation. It was found that exogenously added IL-10 as well as TGF-β1 were able to effectively facilitate LTP evoked with ether high frequency or theta burst stimulation protocols in CA1 area of hippocampus. Effectiveness of IL-10 and TGF-β1 on LTP varied depending on the concentration of used cytokine and type of tetanic stimulation protocol used for LTP induction. Overall the positive effect of studied cytokines on LTP was associated with their ability to increase basal synaptic strength at Schaffer collateral - CA1 synapse. At the same time IL-10 and TGF-β1 did not have any effect on short-term plasticity. Our results provide new evidence upon the modulatory effects that anti-inflammatory cytokines exert on synaptic plasticity further highlighting their potency as modulators of neuronal function.
Collapse
Affiliation(s)
- Miroslav N Nenov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| | - Anton E Malkov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Maxim V Konakov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Sergey G Levin
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
23
|
Warren KN, Beason-Held LL, Carlson O, Egan JM, An Y, Doshi J, Davatzikos C, Ferrucci L, Resnick SM. Elevated Markers of Inflammation Are Associated With Longitudinal Changes in Brain Function in Older Adults. J Gerontol A Biol Sci Med Sci 2019; 73:770-778. [PMID: 29304217 DOI: 10.1093/gerona/glx199] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Indexed: 11/14/2022] Open
Abstract
Background Chronic inflammation has been linked to memory and other cognitive impairments, as well as Alzheimer's disease. Here, we investigate the association between inflammatory markers and changes in brain activity measured by regional cerebral blood flow (rCBF) to assess the relationship between inflammation and brain function in older individuals. Methods Annual 15O water resting-state positron emission tomography (PET) scans collected over a 5-year period were assessed in 138 cognitively normal older participants (77 males; mean age at baseline = 71.3; mean scans per participant = 3.5) in the Baltimore Longitudinal Study of Aging. Voxel-wise linear mixed models were used to investigate associations between rCBF and C-reactive protein (CRP) and interleukin-6 (IL-6) at the time of scanning. We examined relationships between baseline CRP and IL-6 levels and baseline rCBF, and relationships between baseline and mean inflammatory levels over time and longitudinal rCBF changes. Results Higher baseline CRP and IL-6 were each associated with lower baseline rCBF primarily in frontal and occipital regions, with only the lingual gyrus surviving atrophy correction. Higher baseline and mean CRP were also associated with greater rCBF declines over time in anterior cingulate and hippocampal regions, whereas higher baseline and mean IL-6 levels were associated with greater rCBF declines in orbitofrontal and hippocampal regions. Conclusions Higher levels of inflammation are associated with longitudinal changes in brain function in regions important for cognition. These results, along with previous studies, suggest that chronic inflammation in older adults may contribute to age-associated declines in cognitive function.
Collapse
Affiliation(s)
- Kristen N Warren
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| | - Lori L Beason-Held
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| | - Olga Carlson
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| | - Josephine M Egan
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| | - Yang An
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| | - Jimit Doshi
- Department of Radiology, University of Pennsylvania, Philadelphia
| | | | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| | - Susan M Resnick
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland
| |
Collapse
|
24
|
Xiang X, Yu Y, Tang X, Chen M, Zheng Y, Zhu S. Transcriptome Profile in Hippocampus During Acute Inflammatory Response to Surgery: Toward Early Stage of PND. Front Immunol 2019; 10:149. [PMID: 30804943 PMCID: PMC6370675 DOI: 10.3389/fimmu.2019.00149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/17/2019] [Indexed: 01/08/2023] Open
Abstract
Perioperative neurocognitive disorders (PND) are common complications observed in surgical patients, but there are no effective treatments and the detailed mechanisms remain largely unknown. In this study, transcriptome analysis was performed to investigate the hippocampal changes after surgery and underlying molecular mechanisms of PND. Tibial fracture surgery was performed in 3–4 months old C57BL/6J mice to mimic human orthopedic surgery. We demonstrated that memory consolidation of the hippocampal-dependent trace-fear conditioning task was significantly impaired. By using ELISA, a significant elevated IL-6 was observed both in circulating system and central nervous system and peaked at 6 h post-surgery, but transiently returned to baseline thereafter. Hippocampus were collected at 6 h post-surgery then processed for RNA-Seq. A total of 268 genes were screened differentially expressed between the Surgery and Control group, including 170 up-regulated genes and 98 down-regulated genes. By functional enrichment analysis of differently expressed genes, several KEGG pathways involved in inflammatory mediator regulation of TRP channels, neuroactive ligand-receptor interaction and cholinergic synapse were overrepresented. Quantitative real-time PCR confirmed 15 dysregulated genes of interest. These results provide a comprehensive insight into global gene expression changes during the acute presence of hippocampal inflammation and a better understanding on early stage of PND.
Collapse
Affiliation(s)
- Xuwu Xiang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Yu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Tang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Gunn C, Mackus M, Griffin C, Munafò MR, Adams S. A systematic review of the next-day effects of heavy alcohol consumption on cognitive performance. Addiction 2018; 113:2182-2193. [PMID: 30144191 PMCID: PMC6282576 DOI: 10.1111/add.14404] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/03/2018] [Accepted: 07/25/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS Studies examining the next-day cognitive effects of heavy alcohol consumption have produced mixed findings, which may reflect inconsistencies in definitions of 'hangover'. Recent consensus has defined hangover as 'mental and physical symptoms, experienced the day after a single episode of heavy drinking, starting when blood alcohol concentration (BAC) approaches zero'. In light of this, we aimed to review the literature systematically to evaluate and estimate mean effect sizes of the next-day effects of heavy alcohol consumption on cognition. METHODS Embase, PubMed and PsycNET databases were searched between December 2016 and May 2018 using terms based on 'alcohol' and 'hangover'. Studies of experimental designs which reported the next-day cognitive effects of heavy alcohol consumption in a 'hangover' group with BAC < 0.02% were reviewed. A total of 805 articles were identified. Thirty-nine full-text articles were screened by two independent reviewers and 19 included in the systematic review; 11 articles provided sufficient data to be included in the meta-analysis; 1163 participants across 19 studies conducted since 1970 were included in the analysis. Data for study design, hangover severity, BAC at testing and cognitive performance were extracted and effect estimates calculated. RESULTS The systematic review suggested that sustained attention and driving abilities were impaired during hangover. Mixed results were observed for: psychomotor skills, short- (STM) and long-term memory (LTM) and divided attention. The meta-analysis revealed evidence of impairments in STM [g = 0.64, 95% confidence interval (CI) = 0.15-1.13], LTM (Hedges' g = 0.59, 95% CI = 0.01-1.17) sustained attention (g = 0.47, 95% CI = 0.07-0.87) and psychomotor speed (Hedges' g = 0.66, 95% CI = 0.31-1.00) during alcohol hangover. CONCLUSION The research literature suggests that alcohol hangovers may involve impaired cognitive functions and performance of everyday tasks such as driving.
Collapse
Affiliation(s)
- Craig Gunn
- UK Centre for Tobacco and Alcohol Studies and Department of PsychologyUniversity of BathBathUK
| | - Marlou Mackus
- Utrecht Institute for Pharmaceutical Sciences, Department of PsychopharmacologyUtrecht UniversityUtrechtthe Netherlands
| | - Chris Griffin
- UK Centre for Tobacco and Alcohol Studies and Department of PsychologyUniversity of BathBathUK
| | - Marcus R. Munafò
- UK Centre for Tobacco and Alcohol Studies and School of Experimental PsychologyUniversity of BristolBristolUK
- MRC Integrative Epidemiology Unit (IEU) at the University of BristolBristolUK
| | - Sally Adams
- UK Centre for Tobacco and Alcohol Studies and Department of PsychologyUniversity of BathBathUK
| |
Collapse
|
26
|
Bialuk I, Taranta A, Winnicka MM. IL-6 deficiency alters spatial memory in 4- and 24-month-old mice. Neurobiol Learn Mem 2018; 155:21-29. [DOI: 10.1016/j.nlm.2018.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/10/2018] [Accepted: 06/13/2018] [Indexed: 11/26/2022]
|
27
|
Wang TY, Lee SY, Chang YH, Chen SL, Chen PS, Chu CH, Huang SY, Tzeng NS, Lee IH, Chen KC, Yang YK, Chen SH, Hong JS, Lu RB. Correlation of cytokines, BDNF levels, and memory function in patients with opioid use disorder undergoing methadone maintenance treatment. Drug Alcohol Depend 2018; 191:6-13. [PMID: 30071446 PMCID: PMC6487886 DOI: 10.1016/j.drugalcdep.2018.06.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/16/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with opioid use disorder (OUD) show memory deficiencies and impaired treatment outcomes. Emerging evidence suggests that opioid abuse activates proinflammatory processes by increasing cytokine production and impairing neuroprotection, which damages the memory function in OUD patients. Therefore, we investigated whether plasma-based inflammatory and neurotrophic markers correlate with memory function in OUD patients. METHOD OUD patients undergoing methadone maintenance therapy (MMT) were investigated and followed up for 12 weeks. Plasma tumor necrosis factor (TNF)-α, C-reactive protein (CRP), interleukin (IL)-6, transforming growth factor (TGF)-β1, brain-derived neurotrophic factor (BDNF) levels, and Wechsler Memory Scale-Revised (WMS-R) scores were assessed at baseline and after 12 weeks of MMT. Multiple linear regressions and generalized estimating equations (GEEs) were used to examine the correlation between cytokines and memory performance. RESULTS We enrolled 89 patients at baseline; 47 patients completed the end-of-study assessments. Although Pearson correlations showed that CRP and TGF-β1 levels were significantly, negatively associated with some memory indices, the results were not significant after correction. The GEE results, controlled for several confounding factors and multiple testing, showed that changes in TNF-α levels were negatively correlated with changes in the visual memory index (P = 0.01), and that changes in IL-6 levels were negatively correlated with changes in the verbal memory index (P = 0.009). CONCLUSION Memory performance, TNF-α, and IL-6 levels in OUD patients were negative correlated. Additional studies on regulating TNF-α and IL-6 expression to improve memory function in OUD patients might be warranted.
Collapse
Affiliation(s)
- Tzu-Yun Wang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan; Department of Psychiatry, National Cheng Kung University Hospital, Dou-Liou Branch, No. 345, Zhuangjing Road, Douliu, Yunlin 64043, Taiwan.
| | - Sheng-Yu Lee
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Department of Psychiatry, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist., Kaohsiung 81362, Taiwan
| | - Yun-Hsuan Chang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Department of Psychology, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan;,Department of Medical Research, China Medical University Hospital, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Shiou-Lan Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Graduate Institute of Medicine, College of Medicine;,Lipid Science and Aging Research Center, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan
| | - Po-See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Addiction Research Center, National Cheng Kung University, No.1, University Road, Tainan 70101, Taiwan
| | - Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, 3F, No.367, Sheng-Li Rd., North District, Tainan 70456, Taiwan
| | - San-Yuan Huang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei 11490, Taiwan
| | - Nian-Sheng Tzeng
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei 11490, Taiwan;,Student Counseling Center, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei 11490, Taipei, Taiwan
| | - I Hui Lee
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Addiction Research Center, National Cheng Kung University, No.1, University Road, Tainan 70101, Taiwan
| | - Kao Chin Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Addiction Research Center, National Cheng Kung University, No.1, University Road, Tainan 70101, Taiwan
| | - Yen-Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Department of Psychiatry, National Cheng Kung University Hospital, Dou-Liou Branch, No.345, Zhuangjing Rd., Douliu, Yunlin 64043, Taiwan;,Addiction Research Center, National Cheng Kung University, No.1, University Road, Tainan 70101, Taiwan
| | - Shih-Heng Chen
- Neurobiology Laboratory, NIH/NIEHS, Research Triangle Park, 111 T.W. Alexander Drive, Research Triangle Park, N.C. 27709, USA
| | - Jau-Shyong Hong
- Neurobiology Laboratory, NIH/NIEHS, Research Triangle Park, 111 T.W. Alexander Drive, Research Triangle Park, N.C. 27709, USA
| | - Ru-Band Lu
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70428, Taiwan;,Addiction Research Center, National Cheng Kung University, No.1, University Road, Tainan 70101, Taiwan;,Beijing YiNing Hospital, No.9 Minzhuang Road, Haidian District, Beijing 100195, China;,Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
| |
Collapse
|
28
|
Gruol DL, Huitron-Resendiz S, Roberts AJ. Altered brain activity during withdrawal from chronic alcohol is associated with changes in IL-6 signal transduction and GABAergic mechanisms in transgenic mice with increased astrocyte expression of IL-6. Neuropharmacology 2018; 138:32-46. [PMID: 29787738 DOI: 10.1016/j.neuropharm.2018.05.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/25/2018] [Accepted: 05/17/2018] [Indexed: 10/16/2022]
Abstract
Interleukin-6 (IL-6) is an important neuroimmune factor that is increased in the brain by alcohol exposure/withdrawal and is thought to play a role in the actions of alcohol on the brain. To gain insight into IL-6/alcohol/withdrawal interactions and how these interactions affect the brain, we are studying the effects of chronic binge alcohol exposure on transgenic mice that express elevated levels of IL-6 in the brain due to increased astrocyte expression (IL-6 tg) and their non-transgenic (non-tg) littermate controls. IL-6/alcohol/withdrawal interactions were identified by genotypic differences in spontaneous brain activity in electroencephalogram (EEG) recordings from the mice, and by Western blot analysis of protein activation or expression in hippocampus obtained from the mice after the final alcohol withdrawal period. Results from EEG studies showed frequency dependent genotypic differences in brain activity during withdrawal. For EEG frequencies that were affected by alcohol exposure/withdrawal in both genotypes, the nature of the effect was similar, but differed across withdrawal cycles. Differences between IL-6 tg and non-tg mice were also observed in Western blot studies of the activated form of STAT3 (phosphoSTAT3), a signal transduction partner of IL-6, and subunits of GABAA receptors (GABAAR). Regression analysis revealed that pSTAT3 played a more prominent role during withdrawal in the IL-6 tg mice than in the non-tg mice, and that the role of GABAAR alpha-5 and GABAAR alpha-1 in brain activity varied across genotype and withdrawal. Taken together, our results suggest that IL-6 can significantly impact mechanisms involved in alcohol withdrawal.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | | | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
29
|
Bialuk I, Winnicka MM. Facilitatory Effect of IL-6 Deficiency on Long-Term Spatial Memory in Young Adult Mice. Behav Genet 2018; 48:236-246. [DOI: 10.1007/s10519-018-9896-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 03/29/2018] [Indexed: 11/29/2022]
|
30
|
Peineau S, Rabiant K, Pierrefiche O, Potier B. Synaptic plasticity modulation by circulating peptides and metaplasticity: Involvement in Alzheimer's disease. Pharmacol Res 2018; 130:385-401. [PMID: 29425728 DOI: 10.1016/j.phrs.2018.01.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 10/18/2022]
Abstract
Synaptic plasticity is a cellular process involved in learning and memory whose alteration in its two main forms (Long Term Depression (LTD) and Long Term Potentiation (LTP)), is observed in most brain pathologies, including neurodegenerative disorders such as Alzheimer's disease (AD). In humans, AD is associated at the cellular level with neuropathological lesions composed of extracellular deposits of β-amyloid (Aβ) protein aggregates and intracellular neurofibrillary tangles, cellular loss, neuroinflammation and a general brain homeostasis dysregulation. Thus, a dramatic synaptic environment perturbation is observed in AD patients, involving changes in brain neuropeptides, cytokines, growth factors or chemokines concentration and diffusion. Studies performed in animal models demonstrate that these circulating peptides strongly affect synaptic functions and in particular synaptic plasticity. Besides this neuromodulatory action of circulating peptides, other synaptic plasticity regulation mechanisms such as metaplasticity are altered in AD animal models. Here, we will review new insights into the study of synaptic plasticity regulatory/modulatory mechanisms which could influence the process of synaptic plasticity in the context of AD with a particular attention to the role of metaplasticity and peptide dependent neuromodulation.
Collapse
Affiliation(s)
- Stéphane Peineau
- GRAP UMR1247, INSERM, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France; Centre for Synaptic Plasticity, School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK.
| | - Kevin Rabiant
- GRAP UMR1247, INSERM, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Olivier Pierrefiche
- GRAP UMR1247, INSERM, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France.
| | - Brigitte Potier
- Laboratoire Aimé Cotton, CNRS-ENS UMR9188, Université Paris-Sud, Orsay, France.
| |
Collapse
|
31
|
Hu J, Feng X, Valdearcos M, Lutrin D, Uchida Y, Koliwad SK, Maze M. Interleukin-6 is both necessary and sufficient to produce perioperative neurocognitive disorder in mice. Br J Anaesth 2018; 120:537-545. [PMID: 29452810 DOI: 10.1016/j.bja.2017.11.096] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/20/2017] [Accepted: 11/05/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND) result in long-term morbidity and mortality with no effective interventions available. Because interleukin-6 (IL-6), a pro-inflammatory cytokine, is consistently up-regulated by trauma, including after surgery, we determined whether IL-6 is a putative therapeutic target for PND in a mouse model. METHODS Following institutional approval, adult (12-14 weeks) male C57/Bl6 mice were pretreated with the IL-6 receptor (IL6R) blocking antibody tocilizumab prior to open tibia fracture with internal fixation under isoflurane anaesthesia. Inflammatory and behavioural responses in a trace fear conditioning (TFC) paradigm were assessed postoperatively. Separately, the effects of IL-6 administration or of depletion of bone marrow-derived monocytes (BM-DMs) with clodrolip on the inflammatory and behavioural responses were assessed. Blood brain barrier disruption, hippocampal microglial activation, and infiltration of BM-DMs were each assessed following IL-6 administration. RESULTS The surgery-induced decrement in freezing time in the TFC assay, indicative of cognitive decline, was attenuated by tocilizumab (P<0.01). The surgery-induced increase in pro-inflammatory mediators was significantly reduced by tocilizumab. Exogenously administered IL-6 significantly impaired freezing behaviour (P<0.05) and up-regulated pro-inflammatory cytokines; both responses were prevented by depletion of BM-DMs. IL-6 disrupted the blood brain barrier, and increased hippocampal activation of microglia and infiltration of BM-DMs. CONCLUSIONS IL-6 is both necessary and sufficient to produce cognitive decline. Following further preclinical testing of its perioperative safety, the IL6R blocker tocilizumab is a candidate for prevention and/or treatment of PND.
Collapse
Affiliation(s)
- J Hu
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco, CA, USA; Department of Anesthesia, Tongling People's Hospital, Tongling, Anhui 244000, People's Republic of China
| | - X Feng
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco, CA, USA
| | - M Valdearcos
- The Diabetes Center, University of California, San Francisco, CA, USA
| | - D Lutrin
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco, CA, USA
| | - Y Uchida
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco, CA, USA
| | - S K Koliwad
- The Diabetes Center, University of California, San Francisco, CA, USA
| | - M Maze
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco, CA, USA.
| |
Collapse
|
32
|
IL-33 Acts to Express Schaffer Collateral/CA1 LTP and Regulate Learning and Memory by Targeting MyD88. Neural Plast 2017; 2017:2531453. [PMID: 29147584 PMCID: PMC5632899 DOI: 10.1155/2017/2531453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 11/18/2022] Open
Abstract
Interleukin-33 (IL-33) is recognized to transmit a signal through a heterodimeric receptor complex ST2/interleukin-1 receptor accessory protein (IL-1RAcP) bearing activation of myeloid differentiation factor 88 (MyD88). High-frequency stimulation to the Schaffer collateral induced long-term potentiation (LTP) in the CA1 region of hippocampal slices from wild-type control mice. Schaffer collateral/CA1 LTP in IL-33-deficient mice was significantly suppressed, which was neutralized by application with IL-33. Similar suppression of the LTP was found with MyD88-deficient mice but not with ST2-deficient mice. In the water maze test, the acquisition latency in IL-33-deficient and MyD88-deficient mice was significantly prolonged as compared with that in wild-type control mice. Moreover, the retention latency in MyD88-deficient mice was markedly prolonged. In contrast, the acquisition and retention latencies in ST2-deficient mice were not affected. Taken together, these results show that IL-33 acts to express Schaffer collateral/CA1 LTP relevant to spatial learning and memory in a MyD88-dependent manner and that the LTP might be expressed through an IL-1R1/IL-1RAcP-MyD88 pathway in the absence of ST2.
Collapse
|
33
|
Olde Engberink A, Hernandez R, de Graan P, Gruol DL. Rapamycin-Sensitive Late-LTP is Enhanced in the Hippocampus of IL-6 Transgenic Mice. Neuroscience 2017; 367:200-210. [PMID: 29104031 DOI: 10.1016/j.neuroscience.2017.10.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/07/2017] [Accepted: 10/27/2017] [Indexed: 12/13/2022]
Abstract
The neuroimmune factor IL-6 has been shown to regulate hippocampal long-term potentiation (LTP), an activity-dependent enhancement of synaptic transmission that plays a central role in memory and learning. This IL-6 action was demonstrated with relatively short IL-6 exposure, and may reflect physiological actions of IL-6. IL-6 is also expressed chronically at elevated levels in the central nervous system (CNS) under pathological conditions such as neurological disorders. Little is known about the effects IL-6 on LTP under such conditions, an issue that we are addressing by electrophysiological recordings from CA1 pyramidal neurons of hippocampal slices from transgenic mice that persistently express elevated levels of IL-6 in the CNS (IL-6 tg). The current studies examined the long-lasting phase of LTP (late LTP; L-LTP) and the potential involvement mammalian target of rapamycin (mTOR), a known regulator of L-LTP and a downstream partner of IL-6 signal transduction pathways. Results show that basal synaptic transmission and L-LTP were increased in hippocampal slices from IL-6 tg mice compared to slices from non-transgenic (non-tg) control mice. An inhibitor of mTOR, rapamycin, reduced L-LTP in slices from both genotypes, and eliminated the difference in magnitude of L-LTP between IL-6 and non-tg hippocampus. There were no genotypic effect of rapamycin on basal synaptic transmission, but synaptic responses during the LTP induction protocol were reduced in IL-6 tg slices, an effect that could contribute to the reduction of L-LTP in the IL-6 tg slices. These results indicate that persistently increased levels of IL-6 can lead to alterations in mTOR regulation of L-LTP, possibly affecting learning and memory.
Collapse
Affiliation(s)
- Anneke Olde Engberink
- Neuroscience Department, The Scripps Research Institute, San Diego, USA; Department of Translational Neuroscience, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, Netherlands
| | - Ruben Hernandez
- Neuroscience Department, The Scripps Research Institute, San Diego, USA
| | - Pierre de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, Netherlands
| | - Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, San Diego, USA.
| |
Collapse
|
34
|
Wohleb ES, Delpech JC. Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:40-48. [PMID: 27154755 DOI: 10.1016/j.pnpbp.2016.04.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/17/2016] [Accepted: 04/26/2016] [Indexed: 12/15/2022]
Abstract
Psychological stress promotes the development and recurrence of anxiety and depressive behavioral symptoms. Basic and clinical research indicates that stress exposure can influence the neurobiology of mental health disorders through dysregulation of neuroimmune systems. Consistent with this idea several studies show that repeated stress exposure causes microglia activation and recruitment of peripheral monocytes to the brain contributing to development of anxiety- and depressive-like behavior. Further studies show that stress-induced re-distribution of peripheral monocytes leads to stress-sensitized neuroimmune responses and recurrent anxiety-like behavior. These stress-associated immune changes are important because brain resident and peripheral immune cells contribute to physiological processes that support neuroplasticity. Thus, perturbations in neuroimmune function can lead to impaired neuronal responses and synaptic plasticity deficits that underlie behavioral symptoms of mental health disorders. In this review we discuss recent advances in neuroimmune regulation of behavior and summarize studies showing that stress-induced microglia activation and monocyte trafficking in the brain contribute to the neurobiology of mental health disorders.
Collapse
Affiliation(s)
- Eric S Wohleb
- Department of Psychiatry, Yale University School of Medicine, USA.
| | | |
Collapse
|
35
|
Katagiri R, Ishihara-Hattori K, Frings W, Amano J, Fuchs A, Chiba S. Effects of SA237, a humanized anti-interleukin-6 receptor monoclonal antibody, on pre- and postnatal development in cynomolgus monkey. Birth Defects Res 2017; 109:843-856. [DOI: 10.1002/bdr2.1036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Ryuichi Katagiri
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| | | | - Werner Frings
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| | - Jun Amano
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| | - Antje Fuchs
- Covance Preclinical Services GmbH; Muenster Germany
| | - Shuichi Chiba
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| |
Collapse
|
36
|
Onufriev MV, Freiman SV, Peregud DI, Kudryashova IV, Tishkina AO, Stepanichev MY, Gulyaeva NV. Neonatal Proinflammatory Stress Induces Accumulation of Corticosterone and Interleukin-6 in the Hippocampus of Juvenile Rats: Potential Mechanism of Synaptic Plasticity Impairments. BIOCHEMISTRY (MOSCOW) 2017; 82:275-281. [PMID: 28320268 DOI: 10.1134/s0006297917030051] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Infectious diseases in early postnatal ontogenesis can induce neuroinflammation, disrupt normal central nervous system development, and contribute to pathogenesis of cerebral pathologies in adults. To study long-term consequences of such early stress, we induced neonatal proinflammatory stress (NPS) by injecting bacterial lipopolysaccharide into rat pups on postnatal days 3 and 5 and then assessed the levels of corticosterone, proinflammatory cytokines and their mRNAs, and neurotrophins and their mRNAs in the hippocampus and neocortex of the one-month-old animals. Long-term potentiation (LTP) was studied in hippocampal slices as an index of synaptic plasticity. NPS-induced impairments of LTP were accompanied by the accumulation of corticosterone and IL-6 in the hippocampus. In the neocortex, a decrease in exon IV BDNF mRNA was detected. We suggest that excessive corticosterone delivery to hippocampal receptors and proinflammatory changes persisting during brain maturation are among the principal molecular mechanisms responsible for NPS-induced neuroplasticity impairments.
Collapse
Affiliation(s)
- M V Onufriev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
| | | | | | | | | | | | | |
Collapse
|
37
|
Singh A, Abraham WC. Astrocytes and synaptic plasticity in health and disease. Exp Brain Res 2017; 235:1645-1655. [PMID: 28299411 DOI: 10.1007/s00221-017-4928-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/20/2017] [Indexed: 12/22/2022]
Abstract
Activity-dependent synaptic plasticity phenomena such as long-term potentiation and long-term depression are candidate mechanisms for storing information in the brain. Regulation of synaptic plasticity is critical for healthy cognition and learning and this is provided in part by metaplasticity, which can act to maintain synaptic transmission within a dynamic range and potentially prevent excitotoxicity. Metaplasticity mechanisms also allow neurons to integrate plasticity-associated signals over time. Interestingly, astrocytes appear to be critical for certain forms of synaptic plasticity and metaplasticity mechanisms. Synaptic dysfunction is increasingly viewed as an early feature of AD that is correlated with the severity of cognitive decline, and the development of these pathologies is correlated with a rise in reactive astrocytes. This review focuses on the contributions of astrocytes to synaptic plasticity and metaplasticity in normal tissue, and addresses whether astroglial pathology may lead to aberrant engagement of these mechanisms in neurological diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- A Singh
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
38
|
Rae MG, O'Malley D. Cognitive dysfunction in Duchenne muscular dystrophy: a possible role for neuromodulatory immune molecules. J Neurophysiol 2016; 116:1304-15. [PMID: 27385793 DOI: 10.1152/jn.00248.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/29/2016] [Indexed: 11/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X chromosome-linked disease characterized by progressive physical disability, immobility, and premature death in affected boys. Underlying the devastating symptoms of DMD is the loss of dystrophin, a structural protein that connects the extracellular matrix to the cell cytoskeleton and provides protection against contraction-induced damage in muscle cells, leading to chronic peripheral inflammation. However, dystrophin is also expressed in neurons within specific brain regions, including the hippocampus, a structure associated with learning and memory formation. Linked to this, a subset of boys with DMD exhibit nonprogressing cognitive dysfunction, with deficits in verbal, short-term, and working memory. Furthermore, in the genetically comparable dystrophin-deficient mdx mouse model of DMD, some, but not all, types of learning and memory are deficient, and specific deficits in synaptogenesis and channel clustering at synapses has been noted. Little consideration has been devoted to the cognitive deficits associated with DMD compared with the research conducted into the peripheral effects of dystrophin deficiency. Therefore, this review focuses on what is known about the role of full-length dystrophin (Dp427) in hippocampal neurons. The importance of dystrophin in learning and memory is assessed, and the potential importance that inflammatory mediators, which are chronically elevated in dystrophinopathies, may have on hippocampal function is also evaluated.
Collapse
Affiliation(s)
- Mark G Rae
- Department of Physiology, University College Cork, Cork, Ireland; and
| | - Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland; and APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
39
|
Impact of Increased Astrocyte Expression of IL-6, CCL2 or CXCL10 in Transgenic Mice on Hippocampal Synaptic Function. Brain Sci 2016; 6:brainsci6020019. [PMID: 27322336 PMCID: PMC4931496 DOI: 10.3390/brainsci6020019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 12/18/2022] Open
Abstract
An important aspect of CNS disease and injury is the elevated expression of neuroimmune factors. These factors are thought to contribute to processes ranging from recovery and repair to pathology. The complexity of the CNS and the multitude of neuroimmune factors that are expressed in the CNS during disease and injury is a challenge to an understanding of the consequences of the elevated expression relative to CNS function. One approach to address this issue is the use of transgenic mice that express elevated levels of a specific neuroimmune factor in the CNS by a cell type that normally produces it. This approach can provide basic information about the actions of specific neuroimmune factors and can contribute to an understanding of more complex conditions when multiple neuroimmune factors are expressed. This review summarizes studies using transgenic mice that express elevated levels of IL-6, CCL2 or CXCL10 through increased astrocyte expression. The studies focus on the effects of these neuroimmune factors on synaptic function at the Schaffer collateral to CA1 pyramidal neuron synapse of the hippocampus, a brain region that plays a key role in cognitive function.
Collapse
|
40
|
Provencio JJ, Swank V, Lu H, Brunet S, Baltan S, Khapre RV, Seerapu H, Kokiko-Cochran ON, Lamb BT, Ransohoff RM. Neutrophil depletion after subarachnoid hemorrhage improves memory via NMDA receptors. Brain Behav Immun 2016; 54:233-242. [PMID: 26872422 PMCID: PMC4828315 DOI: 10.1016/j.bbi.2016.02.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/29/2016] [Accepted: 02/08/2016] [Indexed: 01/17/2023] Open
Abstract
Cognitive deficits after aneurysmal subarachnoid hemorrhage (SAH) are common and disabling. Patients who experience delayed deterioration associated with vasospasm are likely to have cognitive deficits, particularly problems with executive function, verbal and spatial memory. Here, we report neurophysiological and pathological mechanisms underlying behavioral deficits in a murine model of SAH. On tests of spatial memory, animals with SAH performed worse than sham animals in the first week and one month after SAH suggesting a prolonged injury. Between three and six days after experimental hemorrhage, mice demonstrated loss of late long-term potentiation (L-LTP) due to dysfunction of the NMDA receptor. Suppression of innate immune cell activation prevents delayed vasospasm after murine SAH. We therefore explored the role of neutrophil-mediated innate inflammation on memory deficits after SAH. Depletion of neutrophils three days after SAH mitigates tissue inflammation, reverses cerebral vasoconstriction in the middle cerebral artery, and rescues L-LTP dysfunction at day 6. Spatial memory deficits in both the short and long-term are improved and associated with a shift of NMDA receptor subunit composition toward a memory sparing phenotype. This work supports further investigating suppression of innate immunity after SAH as a target for preventative therapies in SAH.
Collapse
Affiliation(s)
- Jose Javier Provencio
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA; Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA; Department of Neurology and Neuroscience, Brain Immunology and Glia Center, University of Virginia, PO Box 800394, Charlottesville, VA 22908, USA.
| | - Valerie Swank
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Haiyan Lu
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Sylvain Brunet
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Selva Baltan
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Rohini V Khapre
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Himabindu Seerapu
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Olga N Kokiko-Cochran
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Bruce T Lamb
- Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Richard M Ransohoff
- Neuroinflammation Research Center, Neuroscience, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| |
Collapse
|
41
|
Jamalidoust M, Ravanshad M, Namayandeh M, Zare M, Asaei S, Ziyaeyan M. Construction of AAV-rat-IL4 and Evaluation of its Modulating Effect on Aβ (1-42)-Induced Proinflammatory Cytokines in Primary Microglia and the B92 Cell Line by Quantitative PCR Assay. Jundishapur J Microbiol 2016; 9:e30444. [PMID: 27217922 PMCID: PMC4870549 DOI: 10.5812/jjm.30444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/27/2015] [Accepted: 09/29/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Interleukin-4 (IL-4), as the most prominent anti-inflammatory cytokine, plays an important role in modulating microglial activation and inflammatory responses in Alzheimer's disease (AD), a chronic inflammatory disorder. OBJECTIVES The current study aimed to develop a new recombinant Adeno-associated viral (rAAV) vector that delivers IL-4 and then assess the counterbalancing effect of the new construct along with recombinant IL-4 (rIL-4) protein in in-vitro models of AD. MATERIALS AND METHODS The rAAV-IL4 was originally prepared and then employed along with rIL-4 protein to counter Amyloid β (1-42)-induced proinflammatory cytokines in a primary microglia cell culture and the B92 rat microglia continuous cell line, using relative Real-Time PCR assay. RESULTS Aβ (1-42) stimulated the production of the proinflammatory cytokines IL6, IL1β, TNFα, and IL18 in both the primary microglia cell culture and the B92 cell line. Both the rAAV-IL4 construct and the rIL-4 protein were found to inhibit production of the most important Aβ (1-42)-induced proinflammatory cytokine mRNAs in the two types of cells with different patterns. CONCLUSIONS It seems that the new construct can serve as an appropriate option in the modulation of Aβ-induced proinflammatory cytokine gene expression and microglia activation in patients affected by AD.
Collapse
Affiliation(s)
- Marzieh Jamalidoust
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Mehrdad Ravanshad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
- Corresponding author: Mehrdad Ravanshad, Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran. Tel: +98-2182883836, Fax: +98-2188013030, E-mail:
| | - Mandana Namayandeh
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Maryam Zare
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Sadaf Asaei
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Mazyar Ziyaeyan
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| |
Collapse
|
42
|
Hernandez RV, Puro AC, Manos JC, Huitron-Resendiz S, Reyes KC, Liu K, Vo K, Roberts AJ, Gruol DL. Transgenic mice with increased astrocyte expression of IL-6 show altered effects of acute ethanol on synaptic function. Neuropharmacology 2015; 103:27-43. [PMID: 26707655 DOI: 10.1016/j.neuropharm.2015.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/21/2015] [Accepted: 12/14/2015] [Indexed: 11/29/2022]
Abstract
A growing body of evidence has revealed that resident cells of the central nervous system (CNS), and particularly the glial cells, comprise a neuroimmune system that serves a number of functions in the normal CNS and during adverse conditions. Cells of the neuroimmune system regulate CNS functions through the production of signaling factors, referred to as neuroimmune factors. Recent studies show that ethanol can activate cells of the neuroimmune system, resulting in the elevated production of neuroimmune factors, including the cytokine interleukin-6 (IL-6). Here we analyzed the consequences of this CNS action of ethanol using transgenic mice that express elevated levels of IL-6 through increased astrocyte expression (IL-6-tg) to model the increased IL-6 expression that occurs with ethanol use. Results show that increased IL-6 expression induces neuroadaptive changes that alter the effects of ethanol. In hippocampal slices from non-transgenic (non-tg) littermate control mice, synaptically evoked dendritic field excitatory postsynaptic potential (fEPSP) and somatic population spike (PS) at the Schaffer collateral to CA1 pyramidal neuron synapse were reduced by acute ethanol (20 or 60 mM). In contrast, acute ethanol enhanced the fEPSP and PS in hippocampal slices from IL-6 tg mice. Long-term synaptic plasticity of the fEPSP (i.e., LTP) showed the expected dose-dependent reduction by acute ethanol in non-tg hippocampal slices, whereas LTP in the IL-6 tg hippocampal slices was resistant to this depressive effect of acute ethanol. Consistent with altered effects of acute ethanol on synaptic function in the IL-6 tg mice, EEG recordings showed a higher level of CNS activity in the IL-6 tg mice than in the non-tg mice during the period of withdrawal from an acute high dose of ethanol. These results suggest a potential role for neuroadaptive effects of ethanol-induced astrocyte production of IL-6 as a mediator or modulator of the actions of ethanol on the CNS, including persistent changes in CNS function that contribute to cognitive dysfunction and the development of alcohol dependence.
Collapse
Affiliation(s)
- Ruben V Hernandez
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alana C Puro
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jessica C Manos
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Salvador Huitron-Resendiz
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenneth C Reyes
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kevin Liu
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Khanh Vo
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Amanda J Roberts
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Donna L Gruol
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
43
|
Norris CM, Sompol P, Roberts KN, Ansari M, Scheff SW. Pycnogenol protects CA3-CA1 synaptic function in a rat model of traumatic brain injury. Exp Neurol 2015; 276:5-12. [PMID: 26607913 DOI: 10.1016/j.expneurol.2015.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
Abstract
Pycnogenol (PYC) is a patented mix of bioflavonoids with potent anti-oxidant and anti-inflammatory properties. Previously, we showed that PYC administration to rats within hours after a controlled cortical impact (CCI) injury significantly protects against the loss of several synaptic proteins in the hippocampus. Here, we investigated the effects of PYC on CA3-CA1 synaptic function following CCI. Adult Sprague-Dawley rats received an ipsilateral CCI injury followed 15 min later by intravenous injection of saline vehicle or PYC (10 mg/kg). Hippocampal slices from the injured (ipsilateral) and uninjured (contralateral) hemispheres were prepared at seven and fourteen days post-CCI for electrophysiological analyses of CA3-CA1 synaptic function and induction of long-term depression (LTD). Basal synaptic strength was impaired in slices from the ipsilateral, relative to the contralateral, hemisphere at seven days post-CCI and susceptibility to LTD was enhanced in the ipsilateral hemisphere at both post-injury timepoints. No interhemispheric differences in basal synaptic strength or LTD induction were observed in rats treated with PYC. The results show that PYC preserves synaptic function after CCI and provides further rationale for investigating the use of PYC as a therapeutic in humans suffering from neurotrauma.
Collapse
Affiliation(s)
- Christopher M Norris
- Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States; Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| | - Pradoldej Sompol
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| | - Kelly N Roberts
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| | - Mubeen Ansari
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| | - Stephen W Scheff
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40536, United States; Anatomy and Neurobiology, University of Kentucky, College of Medicine, Lexington, KY 40536, United States.
| |
Collapse
|
44
|
Erta M, Giralt M, Esposito FL, Fernandez-Gayol O, Hidalgo J. Astrocytic IL-6 mediates locomotor activity, exploration, anxiety, learning and social behavior. Horm Behav 2015; 73:64-74. [PMID: 26143620 DOI: 10.1016/j.yhbeh.2015.06.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/07/2015] [Accepted: 06/25/2015] [Indexed: 12/27/2022]
Abstract
Interleukin-6 (IL-6) is a major cytokine in the central nervous system, secreted by different brain cells and with roles in a number of physiological functions. We herewith confirm and expand the importance of astrocytic production of and response to IL-6 by using transgenic mice deficient in astrocytic IL-6 (Ast-IL-6 KO) or in its receptor (Ast-IL-6R KO) in full C57Bl/6 genetic background. A major prosurvival effect of astrocytic IL-6 at early ages was clearly demonstrated. Robust effects were also evident in the control of activity and anxiety in the hole-board and elevated plus-maze, and in spatial learning in the Morris water-maze. The results also suggest an inhibitory role of IL-6 in the mechanism controlling the consolidation of hippocampus-dependent spatial learning. Less robust effects of astrocytic IL-6 system were also observed in despair behavior in the tail suspension test, and social behavior in the dominance and resident-intruder tests. The behavioral phenotype was highly dependent on age and/or sex in some cases. The phenotype of Ast-IL-6R KO mice mimicked only partially that of Ast-IL-6KO mice, which indicates both a role of astrocytes in behavior and the participation of other cells besides astrocytes. No evidences of altered function of the hypothalamic-pituitary-adrenal axis were observed. These results demonstrate that astrocytic IL-6 (acting at least partially in astrocytes) regulates normal behavior in mice.
Collapse
Affiliation(s)
- Maria Erta
- Institute of Neurosciences, and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain.
| | - Mercedes Giralt
- Institute of Neurosciences, and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain.
| | - Flavia Lorena Esposito
- Institute of Neurosciences, and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain.
| | - Olaya Fernandez-Gayol
- Institute of Neurosciences, and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain.
| | - Juan Hidalgo
- Institute of Neurosciences, and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
45
|
Phillips DJ, Savenkova MI, Karatsoreos IN. Environmental disruption of the circadian clock leads to altered sleep and immune responses in mouse. Brain Behav Immun 2015; 47:14-23. [PMID: 25542734 DOI: 10.1016/j.bbi.2014.12.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/26/2014] [Accepted: 12/05/2014] [Indexed: 12/29/2022] Open
Abstract
In mammals, one of the most salient outputs of the circadian (daily) clock is the timing of the sleep-wake cycle. Modern industrialized society has led to a fundamental breakdown in the relationship between our endogenous timekeeping systems and the solar day, disrupting normal circadian rhythms. We have argued that disrupted circadian rhythms could lead to changes in allostatic load, and the capacity of organisms to respond to other environmental challenges. In this set of studies, we apply a model of circadian disruption characterized in our lab in which mice are housed in a 20h long day, with 10h of light and 10h of darkness. We explored the effects of this environmental disruption on sleep patterns, to establish if this model results in marked sleep deprivation. Given the interaction between circadian, sleep, and immune systems, we further probed if our model of circadian disruption also alters the innate immune response to peripheral bacterial endotoxin challenge. Our results demonstrate that this model of circadian disruption does not lead to marked sleep deprivation, but instead affects the timing and quality of sleep. We also show that while circadian disruption does not lead to basal changes in the immune markers we explored, the immune response is affected, both in the brain and the periphery. Together, our findings further strengthen the important role of the circadian timing system in sleep regulation and immune responses, and provide evidence that disrupting the circadian clock increases vulnerability to further environmental stressors, including immunological challenges.
Collapse
Affiliation(s)
- Derrick J Phillips
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Marina I Savenkova
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
46
|
Gruol DL. IL-6 regulation of synaptic function in the CNS. Neuropharmacology 2014; 96:42-54. [PMID: 25445486 DOI: 10.1016/j.neuropharm.2014.10.023] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 12/20/2022]
Abstract
A growing body of evidence supports a role for glial-produced neuroimmune factors, including the cytokine IL-6, in CNS physiology and pathology. CNS expression of IL-6 has been documented in the normal CNS at low levels and at elevated levels in several neurodegenerative or psychiatric disease states as well as in CNS infection and injury. The altered CNS function associated with these conditions raises the possibility that IL-6 has neuronal or synaptic actions. Studies in in vitro and in vivo models confirmed this possibility and showed that IL-6 can regulate a number of important neuronal and synaptic functions including synaptic transmission and synaptic plasticity, an important cellular mechanism of memory and learning. Behavioral studies in animal models provided further evidence of an important role for IL-6 as a regulator of CNS pathways that are critical to cognitive function. This review summarizes studies that have lead to our current state of knowledge. In spite of the progress that has been made, there is a need for a greater understanding of the physiological and pathophysiological actions of IL-6 in the CNS, the mechanisms underlying these actions, conditions that induce production of IL-6 in the CNS and therapeutic strategies that could ameliorate or promote IL-6 actions. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Donna L Gruol
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
47
|
Donzis EJ, Tronson NC. Modulation of learning and memory by cytokines: signaling mechanisms and long term consequences. Neurobiol Learn Mem 2014; 115:68-77. [PMID: 25151944 PMCID: PMC4250287 DOI: 10.1016/j.nlm.2014.08.008] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 11/16/2022]
Abstract
This review describes the role of cytokines and their downstream signaling cascades on the modulation of learning and memory. Immune proteins are required for many key neural processes and dysregulation of these functions by systemic inflammation can result in impairments of memory that persist long after the resolution of inflammation. Recent research has demonstrated that manipulations of individual cytokines can modulate learning, memory, and synaptic plasticity. The many conflicting findings, however, have prevented a clear understanding of the precise role of cytokines in memory. Given the complexity of inflammatory signaling, understanding its modulatory role requires a shift in focus from single cytokines to a network of cytokine interactions and elucidation of the cytokine-dependent intracellular signaling cascades. Finally, we propose that whereas signal transduction and transcription may mediate short-term modulation of memory, long-lasting cellular and molecular mechanisms such as epigenetic modifications and altered neurogenesis may be required for the long lasting impact of inflammation on memory and cognition.
Collapse
Affiliation(s)
- Elissa J Donzis
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
48
|
Gruol DL, Vo K, Bray JG. Increased astrocyte expression of IL-6 or CCL2 in transgenic mice alters levels of hippocampal and cerebellar proteins. Front Cell Neurosci 2014; 8:234. [PMID: 25177271 PMCID: PMC4132577 DOI: 10.3389/fncel.2014.00234] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
Emerging research has identified that neuroimmune factors are produced by cells of the central nervous system (CNS) and play critical roles as regulators of CNS function, directors of neurodevelopment and responders to pathological processes. A wide range of neuroimmune factors are produced by CNS cells, primarily the glial cells, but the role of specific neuroimmune factors and their glial cell sources in CNS biology and pathology have yet to be fully elucidated. We have used transgenic mice that express elevated levels of a specific neuroimmune factor, the cytokine IL-6 or the chemokine CCL2, through genetic modification of astrocyte expression to identify targets of astrocyte produced IL-6 or CCL2 at the protein level. We found that in non-transgenic mice constitutive expression of IL-6 and CCL2 occurs in the two CNS regions studied, the hippocampus and cerebellum, as measured by ELISA. In the CCL2 transgenic mice elevated levels of CCL2 were evident in the hippocampus and cerebellum, whereas in the IL-6 transgenic mice, elevated levels of IL-6 were only evident in the cerebellum. Western blot analysis of the cellular and synaptic proteins in the hippocampus and cerebellum of the transgenic mice showed that the elevated levels of CCL2 or IL-6 resulted in alterations in the levels of specific proteins and that these actions differed for the two neuroimmune factors and for the two brain regions. These results are consistent with cell specific profiles of action for IL-6 and CCL2, actions that may be an important aspect of their respective roles in CNS physiology and pathophysiology.
Collapse
Affiliation(s)
- Donna L Gruol
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Khanh Vo
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Jennifer G Bray
- Department of Biology, University of Wisconsin-Stevens Point Stevens Point, WI, USA
| |
Collapse
|
49
|
Costantini M. Body perception, awareness, and illusions. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2014; 5:551-560. [PMID: 26308744 DOI: 10.1002/wcs.1309] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/30/2014] [Accepted: 07/18/2014] [Indexed: 11/08/2022]
Abstract
UNLABELLED Perceiving a body is a phenomenal experience completely different from experiencing a body as one's own body. Visual presentation of bodies or body parts recruits several occipitotemporal regions in the brain. Are these activations sufficient in order to change the phenomenal status of a body in one's own body? In this paper, I will review consolidated experimental evidence showing that the feeling of owning a body is not limited to the vision of a body, rather it is the result of a complex interaction between interoception, exteroception, and pre-existing body templates. To illustrate this complex interplay, I will take advantage of the so-called bodily illusions, referring to controlled illusory generation of unusual bodily feeling. These feelings include having a supernumerary limb, or lacking an arm, or feeling like you do not really have a body, or feeling that you do not really control a certain part of your body, or that your body is not really yours. In the last 15 years more than 150 empirical studies on body illusions have been published ( SOURCE Pubmed, June 2014). These studies, using different technologies, are largely responsible for contributed our current understanding of bodily self-consciousness. WIREs Cogn Sci 2014, 5:551-560. doi: 10.1002/wcs.1309 For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The author has declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- Marcello Costantini
- Laboratory of Neuropsychology and Cognitive Neuroscience, Department of Neuroscience and Imaging, University G. d'Annunzio, Chieti, Italy.,Institute for Advanced Biomedical Technologies ITAB, University G. d'Annunzio, Chieti, Italy.,Mind, Brain Imaging and Neuroethics, University of Ottawa, Institute of Mental Health Research, Ottawa, ON, Canada
| |
Collapse
|
50
|
Sheridan GK, Wdowicz A, Pickering M, Watters O, Halley P, O'Sullivan NC, Mooney C, O'Connell DJ, O'Connor JJ, Murphy KJ. CX3CL1 is up-regulated in the rat hippocampus during memory-associated synaptic plasticity. Front Cell Neurosci 2014; 8:233. [PMID: 25161610 PMCID: PMC4130185 DOI: 10.3389/fncel.2014.00233] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/25/2014] [Indexed: 11/13/2022] Open
Abstract
Several cytokines and chemokines are now known to play normal physiological roles in the brain where they act as key regulators of communication between neurons, glia, and microglia. In particular, cytokines and chemokines can affect cardinal cellular and molecular processes of hippocampal-dependent long-term memory consolidation including synaptic plasticity, synaptic scaling and neurogenesis. The chemokine, CX3CL1 (fractalkine), has been shown to modulate synaptic transmission and long-term potentiation (LTP) in the CA1 pyramidal cell layer of the hippocampus. Here, we confirm widespread expression of CX3CL1 on mature neurons in the adult rat hippocampus. We report an up-regulation in CX3CL1 protein expression in the CA1, CA3 and dentate gyrus (DG) of the rat hippocampus 2 h after spatial learning in the water maze task. Moreover, the same temporal increase in CX3CL1 was evident following LTP-inducing theta-burst stimulation in the DG. At physiologically relevant concentrations, CX3CL1 inhibited LTP maintenance in the DG. This attenuation in dentate LTP was lost in the presence of GABAA receptor/chloride channel antagonism. CX3CL1 also had opposing actions on glutamate-mediated rise in intracellular calcium in hippocampal organotypic slice cultures in the presence and absence of GABAA receptor/chloride channel blockade. Using primary dissociated hippocampal cultures, we established that CX3CL1 reduces glutamate-mediated intracellular calcium rises in both neurons and glia in a dose dependent manner. In conclusion, CX3CL1 is up-regulated in the hippocampus during a brief temporal window following spatial learning the purpose of which may be to regulate glutamate-mediated neurotransmission tone. Our data supports a possible role for this chemokine in the protective plasticity process of synaptic scaling.
Collapse
Affiliation(s)
- Graham K Sheridan
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland ; Department of Physiology, Development and Neuroscience, University of Cambridge Cambridge, UK
| | - Anita Wdowicz
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - Mark Pickering
- School of Medicine and Medical Science, Health Sciences Centre, University College Dublin Dublin, Ireland
| | - Orla Watters
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - Paul Halley
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - Niamh C O'Sullivan
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - Claire Mooney
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - David J O'Connell
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - John J O'Connor
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| | - Keith J Murphy
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin Dublin, Ireland
| |
Collapse
|