51
|
Jiang J, Van TM, Ganesh T, Dingledine R. Discovery of 2-Piperidinyl Phenyl Benzamides and Trisubstituted Pyrimidines as Positive Allosteric Modulators of the Prostaglandin Receptor EP2. ACS Chem Neurosci 2018; 9:699-707. [PMID: 29292987 PMCID: PMC6318807 DOI: 10.1021/acschemneuro.7b00486] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Prostaglandin E2 (PGE2) via its Gαs-coupled EP2 receptor protects cerebral cortical neurons from excitotoxic and anoxic injury, though EP2 receptor activation can also cause secondary neurotoxicity in chronic inflammation. We performed a high-throughput screen of a library of 292 000 small molecules and identified several compounds that have a 2-piperidinyl phenyl benzamide or trisubstituted pyrimidine core as positive modulators for human EP2 receptor. The most active compounds increased the potency of PGE2 on EP2 receptor 4-5-fold at 20 μM without altering efficacy, indicative of an allosteric mechanism. These compounds did not augment the activity of the other Gαs-coupled PGE2 receptor subtype EP4 and showed neuroprotection against N-methyl-d-aspartate (NMDA)-induced excitotoxicity. These newly developed compounds represent second-generation allosteric potentiators for EP2 receptor and shed light on a promising neuroprotective strategy. They should prove valuable as molecular tools to achieve a better understanding of the dichotomous action of brain EP2 receptor activation.
Collapse
Affiliation(s)
- Jianxiong Jiang
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio 45267, United States
| | - Tri Minh Van
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio 45267, United States
| | - Thota Ganesh
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| | - Raymond Dingledine
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
52
|
Synthesis, Characterization, and Biodistribution of Quantum Dot-Celecoxib Conjugate in Mouse Paw Edema Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3090517. [PMID: 30046374 PMCID: PMC6038454 DOI: 10.1155/2018/3090517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Increased risk of cardiovascular side effects has been reported with many of the drugs in the market, including nonsteroidal anti-inflammatory drugs (NSAIDs). Hence, it is critical to thoroughly evaluate the biodistribution and pharmacokinetic properties of the drugs. Presently nanotechnology in combination with noninvasive imaging techniques such as magnetic resonance imaging (MRI), computed axial tomography (CAT), and positron emission tomography (PET) provides a better estimate of the spatio-temporal distribution of therapeutic molecules. Optical imaging using quantum dot- (QD-) tagged biological macromolecules is emerging as a fast, economical, sensitive, and safer alternative for theranostic purposes. In the present study, we report the nanoconjugates of mercaptopropionic acid- (MPA-) capped CdTe quantum dots (QDs) and Celecoxib for bio-imaging in carrageenan-induced mouse paw edema model of inflammation. QD-Celecoxib conjugates were characterized by fluorescence, FT-IR, NMR, and zeta-potential studies. In vivo imaging of QD-Celecoxib conjugates showed clear localization in the inflamed tissue of mouse paw within 3 h, with a gradual increase reaching a maximum and a later decline. This decrease of fluorescence in the paw region is followed by an increase in urinary bladder region, suggesting the possible excretion of QD-drug conjugates from mice within 24 h.
Collapse
|
53
|
Gong Y, Hewett JA. Maintenance of the Innate Seizure Threshold by Cyclooxygenase-2 is Not Influenced by the Translational Silencer, T-cell Intracellular Antigen-1. Neuroscience 2018; 373:37-51. [PMID: 29337236 DOI: 10.1016/j.neuroscience.2018.01.004] [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: 08/22/2017] [Revised: 12/23/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022]
Abstract
Activity of neuronal cyclooxygenase-2 (COX-2), a primary source of PG synthesis in the normal brain, is enhanced by excitatory neurotransmission and this is thought to be involved in seizure suppression. Results herein showing that the incidence of pentylenetetrazole (PTZ)-induced convulsions is suppressed in transgenic mice overexpressing COX-2 in neurons support this notion. T-cell intracellular antigen-1 (TIA-1) is an mRNA binding protein that is known to bind to COX-2 mRNA and repress its translation in non-neuronal cell types. An examination of the expression profile of TIA-1 protein in the normal brain indicated that it is expressed broadly by neurons, including those that express COX-2. However, whether TIA-1 regulates COX-2 protein levels in neurons is not known. The purpose of this study was to test the possibility that deletion of TIA-1 increases basal COX-2 expression in neurons and consequently raises the seizure threshold. Results demonstrate that neither the basal nor seizure-induced expression profiles of COX-2 were altered in mice lacking a functional TIA-1 gene suggesting that TIA-1 does not contribute to regulation of COX-2 protein expression in neurons. The acute PTZ-induced seizure threshold was also unchanged in mice lacking TIA-1 protein, indicating that this RNA binding protein does not influence the innate seizure threshold. Nevertheless, the results raise the possibility that the level of neuronal COX-2 expression may be a determinant of the innate seizure threshold and suggest that a better understanding of the regulation of COX-2 expression in the brain could provide new insight into the molecular mechanisms that suppress seizure induction.
Collapse
Affiliation(s)
- Yifan Gong
- Program in Neuroscience, Department of Biology, Syracuse University, Syracuse, NY 13244, USA
| | - James A Hewett
- Program in Neuroscience, Department of Biology, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
54
|
Mhillaj E, Morgese MG, Tucci P, Furiano A, Luongo L, Bove M, Maione S, Cuomo V, Schiavone S, Trabace L. Celecoxib Prevents Cognitive Impairment and Neuroinflammation in Soluble Amyloid β-treated Rats. Neuroscience 2018; 372:58-73. [PMID: 29306052 DOI: 10.1016/j.neuroscience.2017.12.046] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/11/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
Recent findings suggest that soluble forms of amyloid-β (sAβ) peptide contribute to synaptic and cognitive dysfunctions in early stages of Alzheimer's disease (AD). On the other hand, neuroinflammation and cyclooxygenase-2 (COX-2) enzyme have gained increased interest as key factors involved early in AD, although the signaling pathways and pathophysiologic mechanisms underlying a link between sAβ-induced neurotoxicity and inflammation are still unclear. Here, we investigated the effects of selective COX-2 enzyme inhibition on neuropathological alterations induced by sAβ administration in rats. To this purpose, animals received an intracerebroventricular (icv) injection of predominantly monomeric forms of sAβ and, 7 days after, behavioral as well as biochemical parameters and neurotransmitter alterations were evaluated. During this period, rats also received a sub-chronic treatment with celecoxib. Biochemical results demonstrated that icv sAβ injection significantly increased both COX-2 and pro-inflammatory cytokines expression in the hippocampus (Hipp) of treated rats. In addition, the number of hypertrophic microglial cells and astrocytes were upregulated in sAβ-treated group. Interestingly, rats treated with sAβ showed long-term memory deficits, as confirmed by a significant reduction of discrimination index in the novel object recognition test, along with reduced brain-derived neurotrophic factor expression and increased noradrenaline levels in the Hipp. Systemic administration of celecoxib prevented behavioral dysfunctions, as well as biochemical and neurotransmitter alterations. In conclusion, our results suggest that sAβ neurotoxicity might be associated to COX-2-mediated inflammatory pathways and that early treatment with selective COX-2 inhibitor might provide potential remedies to counterbalance the sAβ-induced effects.
Collapse
Affiliation(s)
- Emanuela Mhillaj
- Dept. of Physiology and Pharmacology, "Sapienza" University of Rome, Rome, Italy
| | - Maria Grazia Morgese
- Dept. of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Paolo Tucci
- Dept. of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Anna Furiano
- Dept. of Experimental Medicine, The Second University of Naples, Naples, Italy
| | - Livio Luongo
- Dept. of Experimental Medicine, The Second University of Naples, Naples, Italy
| | - Maria Bove
- Dept. of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Sabatino Maione
- Dept. of Experimental Medicine, The Second University of Naples, Naples, Italy
| | - Vincenzo Cuomo
- Dept. of Physiology and Pharmacology, "Sapienza" University of Rome, Rome, Italy
| | - Stefania Schiavone
- Dept. of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luigia Trabace
- Dept. of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
55
|
Li B, Guo L, Ku T, Chen M, Li G, Sang N. PM 2.5 exposure stimulates COX-2-mediated excitatory synaptic transmission via ROS-NF-κB pathway. CHEMOSPHERE 2018; 190:124-134. [PMID: 28987401 DOI: 10.1016/j.chemosphere.2017.09.098] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/14/2017] [Accepted: 09/20/2017] [Indexed: 06/07/2023]
Abstract
Long-term exposure to fine particulate matter (PM2.5) has been reported to be closely associated with the neuroinflammation and synaptic dysfunction, but the mechanisms underlying the process remain unclear. Cyclooxygenase-2 (COX-2) is a key player in neuroinflammation, and has been also implicated in the glutamatergic excitotoxicity and synaptic plasticity. Thus, we hypothesized that COX-2 was involved in PM2.5-promoted neuroinflammation and synaptic dysfunction. Our results revealed that PM2.5 elevated COX-2 expression in primary cultured hippocampal neurons and increased the amplitude of field excitatory postsynaptic potentials (fEPSPs) in hippocampal brain slices. And the administration of NS398 (a COX-2 inhibitor) prevented the increased fEPSPs. PM2.5 also induced intracellular reactive oxygen species (ROS) generation accompanied with glutathione (GSH) depletion and the loss of mitochondrial membrane potential (MMP), and the ROS inhibitor, N-acetyl-L-cystein (NAC) suppressed the COX-2 overexpression and the increased fEPSPs. Furthermore, the nuclear factor kappa B (NF-κB) was involved in ROS-induced COX-2 and fEPSP in response to PM2.5 exposure. These findings indicated that PM2.5 activated COX-2 expression and enhanced the synaptic transmission through ROS-NF-κB pathway, and provided possible biomarkers and specific interventions for PM2.5-induced neurological damage.
Collapse
Affiliation(s)
- Ben Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Lin Guo
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Minjun Chen
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
56
|
Rai-Bhogal R, Ahmad E, Li H, Crawford DA. Microarray analysis of gene expression in the cyclooxygenase knockout mice - a connection to autism spectrum disorder. Eur J Neurosci 2017; 47:750-766. [PMID: 29161772 DOI: 10.1111/ejn.13781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
The cellular and molecular events that take place during brain development play an important role in governing function of the mature brain. Lipid-signalling molecules such as prostaglandin E2 (PGE2 ) play an important role in healthy brain development. Abnormalities along the COX-PGE2 signalling pathway due to genetic or environmental causes have been linked to autism spectrum disorder (ASD). This study aims to evaluate the effect of altered COX-PGE2 signalling on development and function of the prenatal brain using male mice lacking cyclooxygenase-1 and cyclooxygenase-2 (COX-1-/- and COX-2-/- ) as potential model systems of ASD. Microarray analysis was used to determine global changes in gene expression during embryonic days 16 (E16) and 19 (E19). Gene Ontology: Biological Process (GO:BP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were implemented to identify affected developmental genes and cellular processes. We found that in both knockouts the brain at E16 had nearly twice as many differentially expressed genes, and affected biological pathways containing various ASD-associated genes important in neuronal function. Interestingly, using GeneMANIA and Cytoscape we also show that the ASD-risk genes identified in both COX-1-/- and COX-2-/- models belong to protein-interaction networks important for brain development despite of different cellular localization of these enzymes. Lastly, we identified eight genes that belong to the Wnt signalling pathways exclusively in the COX-2-/- mice at E16. The level of PKA-phosphorylated β-catenin (S552), a major activator of the Wnt pathway, was increased in this model, suggesting crosstalk between the COX-2-PGE2 and Wnt pathways during early brain development. Overall, these results provide further molecular insight into the contribution of the COX-PGE2 pathways to ASD and demonstrate that COX-1-/- and COX-2-/- animals might be suitable new model systems for studying the disorders.
Collapse
Affiliation(s)
- Ravneet Rai-Bhogal
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.,Department of Biology, York University, Toronto, ON, Canada
| | - Eizaaz Ahmad
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.,Department of Biology, York University, Toronto, ON, Canada
| | - Hongyan Li
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Dorota A Crawford
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.,Department of Biology, York University, Toronto, ON, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
57
|
Wang B, Jin X, Kuang X, Tian S. Chronic administration of parecoxib exerts anxiolytic-like and memory enhancing effects and modulates synaptophysin expression in mice. BMC Anesthesiol 2017; 17:152. [PMID: 29132299 PMCID: PMC5684753 DOI: 10.1186/s12871-017-0443-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 11/02/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Previous studies have shown that cyclooxygenase-2, a key enzyme that converts arachidonic acid to prostaglandins, is involved in anxiety and cognitive processes, but few studies have investigated the effects of chronic administration of cyclooxygenase-2 inhibitors on anxiety, learning and memory under normal physiological conditions. The aim of the study was to investigate the effects of chronic administration of parecoxib, a cyclooxygenase-2 inhibitor, on anxiety behavior and memory performance under normal physiological conditions and to explore the possible neural mechanism underlying parecoxib-mediated effects. METHODS Adult male ICR mice were randomly divided into four groups: the control group and three parecoxib groups. Mice received normal saline or parecoxib (2.5, 5.0 or 10 mg/kg) intraperitoneal injection once a day for 21 days, respectively. Elevated plus-maze, novel object recognition and Y maze tests were conducted on day 23, 24 and 26, respectively. Four additional groups that received same drug treatment were used to measure synaptophysin protein levels by western blot and prostaglandin E2 (PGE2) levels by ELISA in the amygdala and hippocampus on day 26. RESULTS Chronic parecoxib exerted an anxiolytic-like effect in the plus-maze test test, and enhanced memory performance in the novel object recognition and Y maze tests. Western blot analysis showed that chronic parecoxib down-regulated synaptophysin levels in the amygdala and up-regulated synaptophysin levels in the hippocampus. ELISA assay showed that chronic parecoxib inhibited PGE2 in the hippocampus but not amygdala. CONCLUSIONS Chronic parecoxib exerts anxiolytic-like and memory enhancing effects, which might be mediated through differential modulation of synaptophysin and PGE2 in the amygdala and hippocampus.
Collapse
Affiliation(s)
- Bo Wang
- Department of Anesthesiology, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Xin Jin
- Department of Anesthesiology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Xin Kuang
- Department of Anesthesiology, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, People's Republic of China.
| | - Shaowen Tian
- Department of Physiology, College of Medicine, University of South China, Hengyang, Hunan, 421001, People's Republic of China.
| |
Collapse
|
58
|
Eslami SM, Ghasemi M, Bahremand T, Momeny M, Gholami M, Sharifzadeh M, Dehpour AR. Involvement of nitrergic system in anticonvulsant effect of zolpidem in lithium-pilocarpine induced status epilepticus: Evaluation of iNOS and COX-2 genes expression. Eur J Pharmacol 2017; 815:454-461. [DOI: 10.1016/j.ejphar.2017.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 01/04/2023]
|
59
|
Zhu K, Hu M, Yuan B, Liu JX, Liu Y. Aspirin attenuates spontaneous recurrent seizures in the chronically epileptic mice. Neurol Res 2017; 39:744-757. [PMID: 28481152 DOI: 10.1080/01616412.2017.1326657] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Neuroinflammatory processes are pathologic hallmarks of both experimental and human epilepsy, and could be implicated in the neuronal hyperexcitability. Aspirin represents one of the non-selective nonsteroidal anti-inflammatory drugs with fewer side effects in long-term application. This study was carried out to assess the anti-epileptic effects of aspirin when administered during the chronic stage of temporal lobe epilepsy [TLE] in mice. The alteration of hippocampal neurogenesis was also examined for raising a possible mechanism underlying the protective effect of anti-inflammatory treatment in the TLE. METHODS Two months after pilocarpine-induced status epilepticus, the chronically epileptic mice were treated with aspirin (20 mg, 60 mg or 80 mg/kg) once a day for 10 weeks. Spontaneous recurrent seizures were monitored by video camera for 2 weeks. To evaluate the profile of hippocampal neurogenesis, the newly generated cells in the dentate gyrus were labeled by the proliferation marker BrdU. The newborn neurons that extended axons to CA3 area were visualized by cholera toxin B subunit retrograde tracing. RESULTS Administration of aspirin with a dosage of 60 mg or 80 mg/kg initiated at 2 months after pilocarpine-induced status epilepticus significantly reduced the frequency and duration of spontaneous recurrent seizures. Aspirin treatment also increased the number of newborn neurons with anatomic integration through improving the survival of the newly generated cells. CONCLUSION Aspirin treatment during the chronic stage of TLE could attenuate the spontaneous recurrent seizures in mice. Promotion of hippocampal neurogenesis and inhibition of COX-PGE2 pathway might partly contribute to this anti-epileptic effect. Highlights • Aspirin attenuates spontaneous recurrent seizures of chronically epileptic mice • Aspirin increases neurogenesis of chronically epileptic hippocampus by improving the survival of newly generated cells • Promotion of hippocampal neurogenesis and inhibition of COX-PGE2 pathway might partly contribute to anti-epileptic effects of aspirin.
Collapse
Affiliation(s)
- Kun Zhu
- a Institute of Neurobiology , School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center , Xi'an , China
| | - Ming Hu
- a Institute of Neurobiology , School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center , Xi'an , China.,b Department of Human Anatomy, Histology and Embryology , School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center , Xi'an , China
| | - Bo Yuan
- a Institute of Neurobiology , School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center , Xi'an , China
| | - Jian-Xin Liu
- a Institute of Neurobiology , School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center , Xi'an , China
| | - Yong Liu
- a Institute of Neurobiology , School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center , Xi'an , China
| |
Collapse
|
60
|
Patel S, Hill MN, Cheer JF, Wotjak CT, Holmes A. The endocannabinoid system as a target for novel anxiolytic drugs. Neurosci Biobehav Rev 2017; 76:56-66. [PMID: 28434588 PMCID: PMC5407316 DOI: 10.1016/j.neubiorev.2016.12.033] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/22/2016] [Accepted: 12/16/2016] [Indexed: 12/01/2022]
Abstract
The endocannabinoid (eCB) system has attracted attention for its role in various behavioral and brain functions, and as a therapeutic target in neuropsychiatric disease states, including anxiety disorders and other conditions resulting from dysfunctional responses to stress. In this mini-review, we highlight components of the eCB system that offer potential 'druggable' targets for new anxiolytic medications, emphasizing some of the less well-discussed options. We discuss how selectively amplifying eCBs recruitment by interfering with eCB-degradation, via fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), has been linked to reductions in anxiety-like behaviors in rodents and variation in human anxiety symptoms. We also discuss a non-canonical route to regulate eCB degradation that involves interfering with cyclooxygenase-2 (COX-2). Next, we discuss approaches to targeting eCB receptor-signaling in ways that do not involve the cannabinoid receptor subtype 1 (CB1R); by targeting the CB2R subtype and the transient receptor potential vanilloid type 1 (TRPV1). Finally, we review evidence that cannabidiol (CBD), while representing a less specific pharmacological approach, may be another way to modulate eCBs and interacting neurotransmitter systems to alleviate anxiety. Taken together, these various approaches provide a range of plausible paths to developing novel compounds that could prove useful for treating trauma-related and anxiety disorders.
Collapse
Affiliation(s)
- Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, USA; Vanderbilt Kennedy Center for Human Development, Vanderbilt University Medical Center, Nashville, USA
| | - Mathew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada; Departments of Cell Biology and Anatomy and Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology and Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology & Neurogenetics, Munich, Germany
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
61
|
Tripathi P, Singh A, Bala L, Patel DK, Singh MP. Ibuprofen Protects from Cypermethrin-Induced Changes in the Striatal Dendritic Length and Spine Density. Mol Neurobiol 2017; 55:2333-2339. [DOI: 10.1007/s12035-017-0491-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/14/2017] [Indexed: 11/24/2022]
|
62
|
Xu K, Sun X, Benderro GF, Tsipis CP, LaManna JC. Gender differences in hypoxic acclimatization in cyclooxygenase-2-deficient mice. Physiol Rep 2017; 5:5/4/e13148. [PMID: 28242826 PMCID: PMC5328777 DOI: 10.14814/phy2.13148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 01/02/2023] Open
Abstract
The aim of this study was to determine the effect of cyclooxygenase‐2 (COX‐2) gene deletion on the adaptive responses during prolonged moderate hypobaric hypoxia. Wild‐type (WT) and COX‐2 knockout (KO) mice of both genders (3 months old) were exposed to hypobaric hypoxia (~0.4 ATM) or normoxia for 21 days and brain capillary densities were determined. Hematocrit was measured at different time intervals; brain hypoxia‐inducible factor ‐1α (HIF‐1α), angiopoietin 2 (Ang‐2), brain erythropoietin (EPO), and kidney EPO were measured under normoxic and hypoxic conditions. There were no gender differences in hypoxic acclimatization in the WT mice and similar adaptive responses were observed in the female KO mice. However, the male KO mice exhibited progressive vulnerability to prolonged hypoxia. Compared to the WT and female KO mice, the male COX‐2 KO mice had significantly lower survival rate and decreased erythropoietic and polycythemic responses, diminished cerebral angiogenesis, decreased brain accumulation of HIF‐1α, and attenuated upregulation of VEGF, EPO, and Ang‐2 during hypoxia. Our data suggest that there are physiologically important gender differences in hypoxic acclimatization in COX‐2‐deficient mice. The COX‐2 signaling pathway appears to be required for acclimatization in oxygen‐limiting environments only in males, whereas female COX‐2‐deficient mice may be able to access COX‐2‐independent mechanisms to achieve hypoxic acclimatization.
Collapse
Affiliation(s)
- Kui Xu
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Xiaoyan Sun
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Girriso F Benderro
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Constantinos P Tsipis
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Joseph C LaManna
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
63
|
Taşkıran M, Taşdemir A, Ayyıldız N. Acute effects of aceclofenac, COX-2 inhibitor, on penicillin-induced epileptiform activity. Brain Res Bull 2016; 130:42-46. [PMID: 28017780 DOI: 10.1016/j.brainresbull.2016.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022]
Abstract
PURPOSE The effects of COX-2 inhibitors on seizure activity are controversial. The aim of the current study was to determine the post-treatment effect of aceclofenac on penicillin-induced experimental epilepsy. METHODS Male Wistar rats were used in all experiments (n=18). The seizure activity was triggered by penicillin (i.c.). Aceclofenac was injected intraperitoneally at doses of 10mg/kg and 20mg/kg. RESULTS Intraperitoneal administration of 10 and 20mg/kg aceclofenac doses, exhibited proconvulsant properties on seizure activity on rats. The mean spike frequency and amplitude of aceclofenac 10mg/kg were 41.89±2.12 spike/min and 0.619±0.094mV, respectively. The mean spike frequency and amplitude of aceclofenac 20mg/kg were 35.26±2.72 spike/min and 0.843±0.089mV, respectively. CONCLUSION The results indicated that not all of the COX-2 inhibitors may have anticonvulsant or proconvulsant features on patients with epilepsy susceptibility and must be used with great care. It was also suggested that not only cyclooxygenase metabolic pathway but also lipoxygenase pathway should be considered together in further detailed studies.
Collapse
Affiliation(s)
- Mehmet Taşkıran
- Department of Biology, Faculty of Science, Erciyes University, 38039 Kayseri, Turkey.
| | - Abdulkadir Taşdemir
- Graduate School of Natural and Applied Sciences, Erciyes University, 38039 Kayseri, Turkey.
| | - Nusret Ayyıldız
- Department of Biology, Faculty of Science, Erciyes University, 38039 Kayseri, Turkey.
| |
Collapse
|
64
|
Calderón-Garcidueñas L, Maronpot RR, Torres-Jardon R, Henríquez-Roldán C, Schoonhoven R, Acuña-Ayala H, Villarreal-Calderón A, Nakamura J, Fernando R, Reed W, Azzarelli B, Swenberg JA. DNA Damage in Nasal and Brain Tissues of Canines Exposed to Air Pollutants Is Associated with Evidence of Chronic Brain Inflammation and Neurodegeneration. Toxicol Pathol 2016; 31:524-38. [PMID: 14692621 DOI: 10.1080/01926230390226645] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute, subchronic, or chronic exposures to particulate matter (PM) and pollutant gases affect people in urban areas and those exposed to fires, disasters, and wars. Respiratory tract inflammation, production of mediators of inflammation capable of reaching the brain, systemic circulation of PM, and disruption of the nasal respiratory and olfactory barriers are likely in these populations. DNA damage is crucial in aging and in age-associated diseases such as Alzheimer's disease. We evaluated apurinic/apyrimidinic (AP) sites in nasal and brain genomic DNA, and explored by immunohistochemistry the expression of nuclear factor NF κB p65, inducible nitric oxide synthase (iNOS), cyclo-oxygenase 2 (COX2), metallothionein I and II, apolipoprotein E, amyloid precursor protein (APP), and beta-amyloid1-42 in healthy dogs naturally exposed to urban pollution in Mexico City. Nickel (Ni) and vanadium (V) were measured by inductively coupled plasma mass spectrometry (ICP-MS). Forty mongrel dogs, ages 7 days—10 years were studied (14 controls from Tlaxcala and 26 exposed to urban pollution in South West Metropolitan Mexico City (SWMMC)). Nasal respiratory and olfactory epithelium were found to be early pollutant targets. Olfactory bulb and hippocampal AP sites were significantly higher in exposed than in control age matched animals. Ni and V were present in a gradient from olfactory mucosa > olfactory bulb > frontal cortex. Exposed dogs had (a) nuclear neuronal NF κB p65, (b) endothelial, glial and neuronal iNOS, (c) endothelial and glial COX2, (d) ApoE in neuronal, glial and vascular cells, and (e) APP and β amyloid1-42 in neurons, diffuse plaques (the earliest at age 11 months), and in subarachnoid blood vessels. Increased AP sites and the inflammatory and stress protein brain responses were early and significant in dogs exposed to urban pollution. Oil combustion PM-associated metals Ni and V were detected in the brain. There was an acceleration of Alzheimer's-type pathology in dogs chronically exposed to air pollutants. Respiratory tract inflammation and deteriorating olfactory and respiratory barriers may play a role in the observed neuropathology. These data suggest that Alzheimer's disease may be the sequela of air pollutant exposures and the resulting systemic inflammation.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- Environmental Pathology Program, University of North Carolina at Chapel Hill, North Carolina 27599-7310, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Chibowska K, Baranowska-Bosiacka I, Falkowska A, Gutowska I, Goschorska M, Chlubek D. Effect of Lead (Pb) on Inflammatory Processes in the Brain. Int J Mol Sci 2016; 17:ijms17122140. [PMID: 27999370 PMCID: PMC5187940 DOI: 10.3390/ijms17122140] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/10/2016] [Accepted: 12/14/2016] [Indexed: 12/29/2022] Open
Abstract
That the nervous system is the main target of lead (Pb) has long been considered an established fact until recent evidence has linked the Pb effect on the immune system to the toxic effects of Pb on the nervous system. In this paper, we present recent literature reports on the effect of Pb on the inflammatory processes in the brain, particularly the expression of selected cytokines in the brain (interleukin 6, TGF-β1, interleukin 16, interleukin 18, and interleukin 10); expression and activity of enzymes participating in the inflammatory processes, such as cyclooxygenase 2, caspase 1, nitrogen oxide synthase (NOS 2) and proteases (carboxypeptidases, metalloproteinases and chymotrypsin); and the expression of purine receptors P2X4 and P2X7. A significant role in the development of inflammatory processes in the brain is also played by microglia (residual macrophages in the brain and the spinal cord), which act as the first line of defense in the central nervous system, and astrocytes—Whose most important function is to maintain homeostasis for the proper functioning of neurons. In this paper, we also present evidence that exposure to Pb may result in micro and astrogliosis by triggering TLR4-MyD88-NF-κB signaling cascade and the production of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Karina Chibowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| | - Anna Falkowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland.
| | - Marta Goschorska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland.
| |
Collapse
|
66
|
Carta AR, Mulas G, Bortolanza M, Duarte T, Pillai E, Fisone G, Vozari RR, Del-Bel E. l-DOPA-induced dyskinesia and neuroinflammation: do microglia and astrocytes play a role? Eur J Neurosci 2016; 45:73-91. [DOI: 10.1111/ejn.13482] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/07/2016] [Accepted: 11/11/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Anna R. Carta
- Department of Biomedical Sciences; University of Cagliari, Cittadella Universitaria di Monserrato; S.P. N. 8 09042 Monserrato Cagliari Italy
| | - Giovanna Mulas
- Department of Biomedical Sciences; University of Cagliari, Cittadella Universitaria di Monserrato; S.P. N. 8 09042 Monserrato Cagliari Italy
| | - Mariza Bortolanza
- School of Odontology of Ribeirão Preto; Department of Morphology, Physiology and Basic Pathology; University of São Paulo (USP); Av. Café S/N 14040-904 Ribeirão Preto SP Brazil
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
| | - Terence Duarte
- School of Odontology of Ribeirão Preto; Department of Morphology, Physiology and Basic Pathology; University of São Paulo (USP); Av. Café S/N 14040-904 Ribeirão Preto SP Brazil
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
| | - Elisabetta Pillai
- Department of Biomedical Sciences; University of Cagliari, Cittadella Universitaria di Monserrato; S.P. N. 8 09042 Monserrato Cagliari Italy
| | - Gilberto Fisone
- Department of Neuroscience; Karolinska Institutet; Retzius väg 8 17177 Stockholm Sweden
| | - Rita Raisman Vozari
- INSERM U 1127; CNRS UMR 7225; UPMC Univ Paris 06; UMR S 1127; Institut Du Cerveau et de La Moelle Epiniére; ICM; Paris France
| | - Elaine Del-Bel
- School of Odontology of Ribeirão Preto; Department of Morphology, Physiology and Basic Pathology; University of São Paulo (USP); Av. Café S/N 14040-904 Ribeirão Preto SP Brazil
| |
Collapse
|
67
|
Hewett SJ, Shi J, Gong Y, Dhandapani K, Pilbeam C, Hewett JA. Spontaneous Glutamatergic Synaptic Activity Regulates Constitutive COX-2 Expression in Neurons: OPPOSING ROLES FOR THE TRANSCRIPTION FACTORS CREB (cAMP RESPONSE ELEMENT BINDING) PROTEIN AND Sp1 (STIMULATORY PROTEIN-1). J Biol Chem 2016; 291:27279-27288. [PMID: 27875294 DOI: 10.1074/jbc.m116.737353] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 11/10/2016] [Indexed: 11/06/2022] Open
Abstract
Burgeoning evidence supports a role for cyclooxygenase metabolites in regulating membrane excitability in various forms of synaptic plasticity. Two cyclooxygenases, COX-1 and COX-2, catalyze the initial step in the metabolism of arachidonic acid to prostaglandins. COX-2 is generally considered inducible, but in glutamatergic neurons in some brain regions, including the cerebral cortex, it is constitutively expressed. However, the transcriptional mechanisms by which this occurs have not been elucidated. Here, we used quantitative PCR and also analyzed reporter gene expression in a mouse line carrying a construct consisting of a portion of the proximal promoter region of the mouse COX-2 gene upstream of luciferase cDNA to characterize COX-2 basal transcriptional regulation in cortical neurons. Extracts from the whole brain and from the cerebral cortex, hippocampus, and olfactory bulbs exhibited high luciferase activity. Moreover, constitutive COX-2 expression and luciferase activity were detected in cortical neurons, but not in cortical astrocytes, cultured from wild-type and transgenic mice, respectively. Constitutive COX-2 expression depended on spontaneous but not evoked excitatory synaptic activity and was shown to be N-methyl-d-aspartate receptor-dependent. Constitutive promoter activity was reduced in neurons transfected with a dominant-negative cAMP response element binding protein (CREB) and was eliminated by mutating the CRE-binding site on the COX-2 promoter. However, mutation of the stimulatory protein-1 (Sp1)-binding site resulted in an N-methyl-d-aspartate receptor-dependent enhancement of COX-2 promoter activity. Basal binding of the transcription factors CREB and Sp1 to the native neuronal COX-2 promoter was confirmed. In toto, our data suggest that spontaneous glutamatergic synaptic activity regulates constitutive neuronal COX-2 expression via Sp1 and CREB protein-dependent transcriptional mechanisms.
Collapse
Affiliation(s)
- Sandra J Hewett
- From the Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York 13210,
| | - Jingxue Shi
- From the Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York 13210
| | - Yifan Gong
- From the Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York 13210
| | - Krishnan Dhandapani
- the Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, Georgia 30912, and
| | - Carol Pilbeam
- the Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - James A Hewett
- From the Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York 13210,
| |
Collapse
|
68
|
Immunohistochemistry of aberrant neuronal development induced by 6-propyl-2-thiouracil in rats. Toxicol Lett 2016; 261:59-71. [DOI: 10.1016/j.toxlet.2016.08.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 07/03/2016] [Accepted: 08/19/2016] [Indexed: 11/20/2022]
|
69
|
Affiliation(s)
- Douglas A. Kniss
- Department of Obstetrics and Gynecology, Laboratory of Perinatal Research, The Ohio State University, College of Medicine and Public Health, Columbus, Ohio
| |
Collapse
|
70
|
Stacey W, Bhave S, Uht RM. Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression. PLoS One 2016; 11:e0161430. [PMID: 27588681 PMCID: PMC5010190 DOI: 10.1371/journal.pone.0161430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 08/05/2016] [Indexed: 11/19/2022] Open
Abstract
E2 attenuates inflammatory responses by suppressing expression of pro-inflammatory genes. Given that inflammation is increasingly being associated with neurodegenerative and psychiatric processes, we sought to elucidate mechanisms by which E2 down-regulates a component of an inflammatory response, cyclooxygenase- 2 (COX-2) expression. Although inflammatory processes in the brain are usually associated with microglia and astrocytes, we found that the COX-2 gene (cox-2) was expressed in a neuronal context, specifically in an amygdalar cell line (AR-5). Given that COX-2 has been reported to be in neurons in the brain, and that the amygdala is a site involved in neurodegenerative and neuropsychiatric processes, we investigated mechanisms by which E2 could down-regulate cox-2 expression in the AR-5 line. These cells express estrogen receptors alpha (ERα) and beta (ERβ), and as shown here cox-2. At the level of RNA, E2 and the ERβ selective ligand diarylpropionitrile (DPN) both attenuated gene expression, whereas the ERα selective ligand propyl pyrazole triol (PPT) had no effect. Neither ligand increased ERβ at the cox-2 promoter. Rather, DPN decreased promoter occupancy of NF-κB p65 and histone 4 (H4) acetylation. Treatment with the non-specific HDAC inhibitor Trichostatin A (TSA) counteracted DPN's repressive effects on cox-2 expression. In keeping with the TSA effect, E2 and DPN increased histone deacetylase one (HDAC1) and switch-independent 3A (Sin3A) promoter occupancy. Lastly, even though E2 increased CpG methylation, DPN did not. Taken together, the pharmacological data indicate that ERβ contributes to neuronal cox-2 expression, as measured by RNA levels. Furthermore, ER ligands lead to increased recruitment of HDAC1, Sin3A and a concomitant reduction of p65 occupancy and Ac-H4 levels. None of the events, however, are associated with a significant recruitment of ERβ at the promoter. Thus, ERβ directs recruitment to the cox-2 promoter, but does so in the absence of being recruited itself.
Collapse
Affiliation(s)
- Winfred Stacey
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Healthy Aging, Center for Alzheimer’s and Neurodegenerative Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Shreyas Bhave
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Healthy Aging, Center for Alzheimer’s and Neurodegenerative Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Rosalie M. Uht
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Healthy Aging, Center for Alzheimer’s and Neurodegenerative Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- * E-mail:
| |
Collapse
|
71
|
Macht VA. Neuro-immune interactions across development: A look at glutamate in the prefrontal cortex. Neurosci Biobehav Rev 2016; 71:267-280. [PMID: 27593444 DOI: 10.1016/j.neubiorev.2016.08.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 07/26/2016] [Accepted: 08/31/2016] [Indexed: 02/07/2023]
Abstract
Although the primary role for the immune system is to respond to pathogens, more recently, the immune system has been demonstrated to have a critical role in signaling developmental events. Of particular interest for this review is how immunocompetent microglia and astrocytes interact with glutamatergic systems to influence the development of neural circuits in the prefrontal cortex (PFC). Microglia are the resident macrophages of the brain, and astrocytes mediate both glutamatergic uptake and coordinate with microglia to respond to the general excitatory state of the brain. Cross-talk between microglia, astrocytes, and glutamatergic neurons forms a quad-partite synapse, and this review argues that interactions within this synapse have critical implications for the maturation of PFC-dependent cognitive function. Similarly, understanding developmental shifts in immune signaling may help elucidate variations in sensitivities to developmental disruptions.
Collapse
Affiliation(s)
- Victoria A Macht
- University of South Carolina, 1512 Pendleton St., Department of Psychology, Columbia, SC 29208, United States.
| |
Collapse
|
72
|
Meta-analysis of cyclooxygenase-2 (COX-2) 765G>C polymorphism and Alzheimer’s disease. J Clin Neurosci 2016; 31:4-9. [DOI: 10.1016/j.jocn.2015.11.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/30/2015] [Indexed: 01/19/2023]
|
73
|
Musto AE, Rosencrans RF, Walker CP, Bhattacharjee S, Raulji CM, Belayev L, Fang Z, Gordon WC, Bazan NG. Dysfunctional epileptic neuronal circuits and dysmorphic dendritic spines are mitigated by platelet-activating factor receptor antagonism. Sci Rep 2016; 6:30298. [PMID: 27444269 PMCID: PMC4957208 DOI: 10.1038/srep30298] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/30/2016] [Indexed: 01/28/2023] Open
Abstract
Temporal lobe epilepsy or limbic epilepsy lacks effective therapies due to a void in understanding the cellular and molecular mechanisms that set in motion aberrant neuronal network formations during the course of limbic epileptogenesis (LE). Here we show in in vivo rodent models of LE that the phospholipid mediator platelet-activating factor (PAF) increases in LE and that PAF receptor (PAF-r) ablation mitigates its progression. Synthetic PAF-r antagonists, when administered intraperitoneally in LE, re-establish hippocampal dendritic spine density and prevent formation of dysmorphic dendritic spines. Concomitantly, hippocampal interictal spikes, aberrant oscillations, and neuronal hyper-excitability, evaluated 15–16 weeks after LE using multi-array silicon probe electrodes implanted in the dorsal hippocampus, are reduced in PAF-r antagonist-treated mice. We suggest that over-activation of PAF-r signaling induces aberrant neuronal plasticity in LE and leads to chronic dysfunctional neuronal circuitry that mediates epilepsy.
Collapse
Affiliation(s)
- Alberto E Musto
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Robert F Rosencrans
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Chelsey P Walker
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Chittalsinh M Raulji
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA.,Department of Pediatrics, Hematology-Oncology, Louisiana State University Health Sciences Center and Children's Hospital of New Orleans, New Orleans, Louisiana 70118, USA
| | - Ludmila Belayev
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Zhide Fang
- Biostatistics, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - William C Gordon
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| |
Collapse
|
74
|
De Felice A, Greco A, Calamandrei G, Minghetti L. Prenatal exposure to the organophosphate insecticide chlorpyrifos enhances brain oxidative stress and prostaglandin E2 synthesis in a mouse model of idiopathic autism. J Neuroinflammation 2016; 13:149. [PMID: 27301868 PMCID: PMC4908699 DOI: 10.1186/s12974-016-0617-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) are emerging as polygenic and multifactorial disorders in which complex interactions between defective genes and early exposure to environmental stressors impact on the correct neurodevelopment and brain processes. Organophosphate insecticides, among which chlorpyrifos (CPF), are widely diffused environmental toxicants associated with neurobehavioral deficits and increased risk of ASD occurrence in children. Oxidative stress and dysregulated immune responses are implicated in both organophosphate neurodevelopmental effects and ASD etiopathogenesis. BTBR T+tf/J mice, a well-studied model of idiopathic autism, show several behavioral and immunological alterations found in ASD children, and we recently showed that CPF gestational exposure strengthened some of these autistic-like traits. In the present study, we aimed at investigating whether the behavioral effects of gestational CPF administration are associated with brain increased oxidative stress and altered lipid mediator profile. METHODS Brain levels of F2-isoprostanes (15-F2t-IsoP), as index of in vivo oxidative stress, and prostaglandin E2 (PGE2), a major arachidonic acid metabolite released by immune cells and by specific glutamatergic neuron populations mainly in cortex and hippocampus, were assessed by specific enzyme-immuno assays in brain homogenates from BTBR T+tf/J and C57Bl6/J mice, exposed during gestation to either vehicle or CPF. Measures were performed in mice of both sexes, at different postnatal stages (PNDs 1, 21, and 70). RESULTS At birth, BTBR T+tf/J mice exhibited higher baseline 15-F2t-IsoP levels as compared to C57Bl6/J mice, suggestive of greater oxidative stress processes. Gestational treatment with CPF-enhanced 15-F2t-IsoP and PGE2 levels in strain- and age-dependent manner, with 15-F2t-IsoP increased in BTBR T+tf/J mice at PNDs 1 and 21, and PGE2 elevated in BTBR T+tf/J mice at PNDs 21 and 70. At PND 21, CPF effects were sex-dependent being the increase of the two metabolites mainly associated with male mice. CPF treatment also induced a reduction of somatic growth, which reached statistical significance at PND 21. CONCLUSIONS These findings indicate that the autistic-like BTBR T+tf/J strain is highly vulnerable to environmental stressors during gestational period. The results further support the hypothesis that oxidative stress might be the link between environmental neurotoxicants such as CPF and ASD. The increased levels of oxidative stress during early postnatal life could result in delayed and long-lasting alterations in specific pathways relevant to ASD, of which PGE2 signaling represents an important one.
Collapse
Affiliation(s)
- Alessia De Felice
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy
- Present address: Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Via Bettini 31, 38068, Rovereto (TN), Italy
| | - Anita Greco
- Section of Experimental Neurology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy
| | - Gemma Calamandrei
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy
| | - Luisa Minghetti
- Section of Experimental Neurology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, I-00161, Rome, Italy.
| |
Collapse
|
75
|
Woodling NS, Colas D, Wang Q, Minhas P, Panchal M, Liang X, Mhatre SD, Brown H, Ko N, Zagol-Ikapitte I, van der Hart M, Khroyan TV, Chuluun B, Priyam PG, Milne GL, Rassoulpour A, Boutaud O, Manning-Boğ AB, Heller HC, Andreasson KI. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer's disease model mice. Brain 2016; 139:2063-81. [PMID: 27190010 DOI: 10.1093/brain/aww117] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/31/2016] [Indexed: 01/22/2023] Open
Abstract
Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-β accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-β oligomers.
Collapse
Affiliation(s)
- Nathaniel S Woodling
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 2 Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Damien Colas
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Qian Wang
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paras Minhas
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maharshi Panchal
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xibin Liang
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Siddhita D Mhatre
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Holden Brown
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 4 Brains On-line LLC, South San Francisco, CA, USA
| | - Novie Ko
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irene Zagol-Ikapitte
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marieke van der Hart
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taline V Khroyan
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bayarsaikhan Chuluun
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Prachi G Priyam
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ginger L Milne
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arash Rassoulpour
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Olivier Boutaud
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amy B Manning-Boğ
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Craig Heller
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katrin I Andreasson
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
76
|
Gamble-George JC, Baldi R, Halladay L, Kocharian A, Hartley N, Silva CG, Roberts H, Haymer A, Marnett LJ, Holmes A, Patel S. Cyclooxygenase-2 inhibition reduces stress-induced affective pathology. eLife 2016; 5:e14137. [PMID: 27162170 PMCID: PMC4862754 DOI: 10.7554/elife.14137] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/09/2016] [Indexed: 12/31/2022] Open
Abstract
Mood and anxiety disorders are the most prevalent psychiatric conditions and are exacerbated by stress. Recent studies have suggested cyclooxygenase-2 (COX-2) inhibition could represent a novel treatment approach or augmentation strategy for affective disorders including anxiety disorders and major depression. We show that traditional COX-2 inhibitors and a newly developed substrate-selective COX-2 inhibitor (SSCI) reduce a variety of stress-induced behavioral pathologies in mice. We found that these behavioral effects were associated with a dampening of neuronal excitability in the basolateral amygdala (BLA) ex vivo and in vivo, and were mediated by small-conductance calcium-activated potassium (SK) channel and CB1 cannabinoid receptor activation. Taken together, these data provide further support for the potential utility of SSCIs, as well as traditional COX-2 inhibitors, as novel treatment approaches for stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Joyonna Carrie Gamble-George
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, United States
| | - Rita Baldi
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
| | - Lindsay Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse, Bethesda, United States
| | - Adrina Kocharian
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse, Bethesda, United States
| | - Nolan Hartley
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, United States
| | - Carolyn Grace Silva
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
| | - Holly Roberts
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
| | - Andre Haymer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
| | - Lawrence J Marnett
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, and Vanderbilt-Ingram Cancer Center, Nashville, United States
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse, Bethesda, United States
| | - Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, United States
- Vanderbilt Kennedy Center for Human Development, Vanderbilt University Medical Center, Nashville, United States
| |
Collapse
|
77
|
Du C, Zheng F, Wang X. Exploring novel AEDs from drugs used for treatment of non-epileptic disorders. Expert Rev Neurother 2016; 16:449-61. [PMID: 27010915 DOI: 10.1586/14737175.2016.1158101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Epilepsy is a chronic neurological disease. Although many anti-epileptic drugs (AEDs) have been developed for clinical use, they have no effect on 20-30% of patients and do not generally prevent epileptogenesis. Because of the long development cycle for new AEDs and the high cost, increasing efforts are being made to find anti-epileptic effects among drugs that are already listed for the treatment of other diseases and repurpose them as potential anti-epileptic treatments. Here, we review the progress that has been made in this field as a result of animal and clinical trials of drugs such as rapamycin, everolimus, losartan, celecoxib, bumetanide and other non-epileptic drugs. These drugs can prevent the epileptogenesis, reduce the epileptic pathological changes, and even be used to treat intractable epilepsy. Their mechanisms of action are completely different from those of existing AEDs, prompting researchers to change their perspectives in the search for new AEDs.
Collapse
Affiliation(s)
- Chao Du
- a Department of Neurology , The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Fangshuo Zheng
- a Department of Neurology , The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Xuenfeng Wang
- a Department of Neurology , The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| |
Collapse
|
78
|
Codocedo JF, Inestrosa NC. Wnt-5a-regulated miR-101b controls COX2 expression in hippocampal neurons. Biol Res 2016; 49:9. [PMID: 26895946 PMCID: PMC4759731 DOI: 10.1186/s40659-016-0071-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Wnt-5a is a member of the WNT family of secreted lipoglycoproteins, whose expression increases during development; moreover, Wnt-5a plays a key role in synaptic structure and function in the adult nervous system. However, the mechanism underlying these effects is still elusive. MicroRNAs (miRNAs) are a family of small non-coding RNAs that control the gene expression of their targets through hybridization with complementary sequences in the 3′ UTR, thereby inhibiting the translation of the target proteins. Several evidences indicate that the miRNAs are actively involved in the regulation of neuronal function. Results In the present study, we examined whether Wnt-5a modulates the levels of miRNAs in hippocampal neurons. Using PCR arrays, we identified a set of miRNAs that respond to Wnt-5a treatment. One of the most affected miRNAs was miR-101b, which targets cyclooxygenase-2 (COX2), an inducible enzyme that converts arachidonic acid to prostanoids, and has been involved in the injury/inflammatory response, and more recently in neuronal plasticity. Consistent with the Wnt-5a regulation of miR-101b, this Wnt ligand regulates COX2 expression in a time-dependent manner in cultured hippocampal neurons. Conclusion The biological processes induced by Wnt-5a in hippocampal neurons, involve the regulation of several miRNAs including miR-101b, which has the capacity to regulate several targets, including COX-2 in the central nervous system.
Collapse
Affiliation(s)
- Juan Francisco Codocedo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile. .,CARE, Biomedical Research Center, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile.
| | - Nibaldo C Inestrosa
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile. .,Faculty of Medicine, Center for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,CARE, Biomedical Research Center, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile.
| |
Collapse
|
79
|
Bortolanza M, Padovan-Neto FE, Cavalcanti-Kiwiatkoski R, Dos Santos-Pereira M, Mitkovski M, Raisman-Vozari R, Del-Bel E. Are cyclooxygenase-2 and nitric oxide involved in the dyskinesia of Parkinson's disease induced by L-DOPA? Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0190. [PMID: 26009769 DOI: 10.1098/rstb.2014.0190] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Inflammatory mechanisms are proposed to play a role in L-DOPA-induced dyskinesia. Cyclooxygenase-2 (COX2) contributes to inflammation pathways in the periphery and is constitutively expressed in the central nervous system. Considering that inhibition of nitric oxide (NO) formation attenuates L-DOPA-induced dyskinesia, this study aimed at investigating if a NO synthase (NOS) inhibitor would change COX2 brain expression in animals with L-DOPA-induced dyskinesia. To this aim, male Wistar rats received unilateral 6-hydroxydopamine microinjection into the medial forebrain bundle were treated daily with L-DOPA (21 days) combined with 7-nitroindazole or vehicle. All hemi-Parkinsonian rats receiving l-DOPA showed dyskinesia. They also presented increased neuronal COX2 immunoreactivity in the dopamine-depleted dorsal striatum that was directly correlated with dyskinesia severity. Striatal COX2 co-localized with choline-acetyltransferase, calbindin and DARPP-32 (dopamine-cAMP-regulated phosphoprotein-32), neuronal markers of GABAergic neurons. NOS inhibition prevented L-DOPA-induced dyskinesia and COX2 increased expression in the dorsal striatum. These results suggest that increased COX2 expression after L-DOPA long-term treatment in Parkinsonian-like rats could contribute to the development of dyskinesia.
Collapse
Affiliation(s)
- Mariza Bortolanza
- School of Odontology of Ribeirão Preto, Department of Morphology, University of São Paulo (USP), Physiology and Basic Pathology, Av. Café S/N, 14040-904, Ribeirão Preto, São Paulo, Brazil Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil
| | - Fernando E Padovan-Neto
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Department of Behavioural Neurosciences, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil
| | - Roberta Cavalcanti-Kiwiatkoski
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| | - Maurício Dos Santos-Pereira
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| | - Miso Mitkovski
- Light Microscopy Facility, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Rita Raisman-Vozari
- Institut de Cerveau et de la Moelle Epinière, Sorbonne Université UPMC UM75 INSERM U1127, CNRS UMR 7225, Paris, France
| | - Elaine Del-Bel
- School of Odontology of Ribeirão Preto, Department of Morphology, University of São Paulo (USP), Physiology and Basic Pathology, Av. Café S/N, 14040-904, Ribeirão Preto, São Paulo, Brazil Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Department of Behavioural Neurosciences, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
80
|
Abe H, Saito F, Tanaka T, Mizukami S, Hasegawa-Baba Y, Imatanaka N, Akahori Y, Yoshida T, Shibutani M. Developmental cuprizone exposure impairs oligodendrocyte lineages differentially in cortical and white matter tissues and suppresses glutamatergic neurogenesis signals and synaptic plasticity in the hippocampal dentate gyrus of rats. Toxicol Appl Pharmacol 2016; 290:10-20. [DOI: 10.1016/j.taap.2015.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/10/2015] [Indexed: 10/22/2022]
|
81
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015; 35:11791-810. [PMID: 26311764 DOI: 10.1523/jneurosci.0651-15.2015] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
82
|
Srivastava A, Dixit AB, Banerjee J, Tripathi M, Sarat Chandra P. Role of inflammation and its miRNA based regulation in epilepsy: Implications for therapy. Clin Chim Acta 2015; 452:1-9. [PMID: 26506013 DOI: 10.1016/j.cca.2015.10.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/19/2015] [Accepted: 10/22/2015] [Indexed: 02/06/2023]
Abstract
There is a need to develop innovative therapeutic strategies to counteract epilepsy, a common disabling neurological disorder. Despite the recent advent of additional antiepileptic drugs and respective surgery, the treatment of epilepsy remains a major challenge. The available therapies are largely based on symptoms, and these approaches do not affect the underlying disease processes and are also associated frequently with severe side effects. This is mainly because of the lack of well-defined targets in epilepsy. The discovery that inflammatory mediators significantly contribute to the onset and recurrence of seizures in experimental seizure models, as well as the presence of inflammatory molecules in human epileptogenic tissue, highlights the possibility of targeting specific inflammation related pathways to control seizures that are otherwise resistant to the available AEDs. Emerging studies suggest that miRNAs have a significant role in regulating inflammatory pathways shown to be involved in epilepsy. These miRNAs can possibly be used as novel therapeutic targets in the treatment of epilepsy as well as serve as diagnostic biomarkers of epileptogenesis. This review highlights the immunological features underlying the pathogenesis of epileptic seizures and the possible miRNA mediated approaches for drug resistant epilepsies that modulate the immune-mediated pathogenesis.
Collapse
Affiliation(s)
- Arpna Srivastava
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Aparna Banerjee Dixit
- Center of Excellence for Epilepsy, A joint NBRC-AIIMS collaboration, NBRC, Manesar, India
| | - Jyotirmoy Banerjee
- Center of Excellence for Epilepsy, A joint NBRC-AIIMS collaboration, NBRC, Manesar, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - P Sarat Chandra
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
83
|
Pathophysiological Roles of Cyclooxygenases and Prostaglandins in the Central Nervous System. Mol Neurobiol 2015; 53:4754-71. [PMID: 26328537 DOI: 10.1007/s12035-015-9355-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 07/07/2015] [Indexed: 01/01/2023]
Abstract
Cyclooxygenases (COXs) oxidize arachidonic acid to prostaglandin (PG) G2 and H2 followed by PG synthases that generates PGs and thromboxane (TX) A2. COXs are divided into COX-1 and COX-2. In the central nervous system, COX-1 is constitutively expressed in neurons, astrocytes, and microglial cells. COX-2 is upregulated in these cells under pathophysiological conditions. In hippocampal long-term potentiation, COX-2, PGE synthase, and PGE2 are induced in post-synaptic neurons. PGE2 acts pre-synaptic EP2 receptor, generates cAMP, stimulates protein kinase A, modulates voltage-dependent calcium channel, facilitates glutamatergic synaptic transmission, and potentiates long-term plasticity. PGD2, PGE2, and PGI2 exhibit neuroprotective effects via Gs-coupled DP1, EP2/EP4, and IP receptors, respectively. COX-2, PGD2, PGE2, PGF2α, and TXA2 are elevated in stroke. COX-2 inhibitors exhibit neuroprotective effects in vivo and in vitro models of stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, epilepsy, and schizophrenia, suggesting neurotoxicities of COX products. PGE2, PGF2α, and TXA2 can contribute to the neurodegeneration via EP1, FP, and TP receptors, respectively, which are coupled with Gq, stimulate phospholipase C and cleave phosphatidylinositol diphosphate to produce inositol triphosphate and diacylglycerol. Inositol triphosphate binds to inositol triphosphate receptor in endoplasmic reticulum, releases calcium, and results in increasing intracellular calcium concentrations. Diacylglycerol activates calcium-dependent protein kinases. PGE2 disrupts Ca(2+) homeostasis by impairing Na(+)-Ca(2+) exchange via EP1, resulting in the excess Ca(2+) accumulation. Neither PGE2, PGF2α, nor TXA2 causes neuronal cell death by itself, suggesting that they might enhance the ischemia-induced neurodegeneration. Alternatively, PGE2 is non-enzymatically dehydrated to a cyclopentenone PGA2, which induces neuronal cell death. Although PGD2 induces neuronal apoptosis after a lag time, neither DP1 nor DP2 is involved in the neurotoxicity. As well as PGE2, PGD2 is non-enzymatically dehydrated to a cyclopentenone 15-deoxy-Δ(12,14)-PGJ2, which induces neuronal apoptosis without a lag time. However, neurotoxicities of these cyclopentenones are independent of their receptors. The COX-2 inhibitor inhibits both the anchorage-dependent and anchorage-independent growth of glioma cell lines regardless of COX-2 expression, suggesting that some COX-2-independent mechanisms underlie the antineoplastic effect of the inhibitor. PGE2 attenuates this antineoplastic effect, suggesting that the predominant mechanism is COX-dependent. COX-2 or EP1 inhibitors show anti-neoplastic effects. Thus, our review presents evidences for pathophysiological roles of cyclooxygenases and prostaglandins in the central nervous system.
Collapse
|
84
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015. [PMID: 26311764 DOI: 10.1523/jneurosci.0651‐15.2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
85
|
Desmarais G, Charest G, Fortin D, Bujold R, Mathieu D, Paquette B. Cyclooxygenase-2 inhibitor prevents radiation-enhanced infiltration of F98 glioma cells in brain of Fischer rat. Int J Radiat Biol 2015; 91:624-33. [PMID: 25912457 DOI: 10.3109/09553002.2015.1043756] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Radiation induces a neuro-inflammation that is characterized by the expression of genes known to increase the invasion of cancer cells. In Fischer rats, brain irradiation increases the infiltration of cancer cells and reduced the median survival of the animals. In this study, we have determined whether these adverse effects of radiation can be prevented with the cyclooxygenase-2 (COX-2) inhibitor meloxicam. MATERIALS AND METHODS Brain of Fischer rats treated or not with meloxicam were irradiated (15 Gy) and then implanted with the F98 glioma cells. The median survival of the animals, the infiltration of F98 cells, and the expression of inflammatory cytokines and pro-migration molecules were measured. RESULTS Meloxicam reduced by 75% the production of prostaglandin E2 (bioproduct of COX-2) in irradiated brains validating its anti-inflammatory effect. Median survival was increased to control levels by the treatment of meloxicam following brain irradiation. This protective effect was associated with a reduction of the infiltration of F98 cells in the brain, a complete inhibition of radiation-enhancement of matrix metalloproteinase-2, and a significant reduction of tumor necrosis factor α (TNF-α) and tumor growth factor β1 (TGF-β1) expression. Using invasion chambers, interleukin-1β (IL-1β) stimulated by 5-fold the invasiveness of F98 cells, but this stimulation was completely inhibited by meloxicam. This suggests that a cooperation between IL-1β and COX-2 are involved in radiation-enhancement of F98 cell invasion. CONCLUSIONS Our results indicate the importance of reducing the inflammatory response of normal brain tissue following irradiation in an effort to extend median survival in F98 tumor-bearing rats.
Collapse
Affiliation(s)
- Guillaume Desmarais
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada
| | - Gabriel Charest
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada
| | - David Fortin
- b Department of Surgery , Division of Neurosurgery/Neuro-oncology , Québec , Canada
| | - Rachel Bujold
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada.,c Division of Radiation Oncology, Centre Hospitalier Universitaire de Sherbrooke , Sherbrooke, Québec , Canada
| | - David Mathieu
- b Department of Surgery , Division of Neurosurgery/Neuro-oncology , Québec , Canada
| | - Benoit Paquette
- a Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology , Faculty of Medicine and Health Sciences, Université de Sherbrooke , Québec , Canada
| |
Collapse
|
86
|
Chicca A, Gachet MS, Petrucci V, Schuehly W, Charles RP, Gertsch J. 4'-O-methylhonokiol increases levels of 2-arachidonoyl glycerol in mouse brain via selective inhibition of its COX-2-mediated oxygenation. J Neuroinflammation 2015; 12:89. [PMID: 25962384 PMCID: PMC4490613 DOI: 10.1186/s12974-015-0307-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/24/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE 4'-O-methylhonokiol (MH) is a natural product showing anti-inflammatory, anti-osteoclastogenic, and neuroprotective effects. MH was reported to modulate cannabinoid CB2 receptors as an inverse agonist for cAMP production and an agonist for intracellular [Ca2+]. It was recently shown that MH inhibits cAMP formation via CB2 receptors. In this study, the exact modulation of MH on CB2 receptor activity was elucidated and its endocannabinoid substrate-specific inhibition (SSI) of cyclooxygenase-2 (COX-2) and CNS bioavailability are described for the first time. METHODS CB2 receptor modulation ([35S]GTPγS, cAMP, and β-arrestin) by MH was measured in hCB2-transfected CHO-K1 cells and native conditions (HL60 cells and mouse spleen). The COX-2 SSI was investigated in RAW264.7 cells and in Swiss albino mice by targeted metabolomics using LC-MS/MS. RESULTS MH is a CB2 receptor agonist and a potent COX-2 SSI. It induced partial agonism in both the [35S]GTPγS binding and β-arrestin recruitment assays while being a full agonist in the cAMP pathway. MH selectively inhibited PGE2 glycerol ester formation (over PGE2) in RAW264.7 cells and significantly increased the levels of 2-AG in mouse brain in a dose-dependent manner (3 to 20 mg kg(-1)) without affecting other metabolites. After 7 h from intraperitoneal (i.p.) injection, MH was quantified in significant amounts in the brain (corresponding to 200 to 300 nM). CONCLUSIONS LC-MS/MS quantification shows that MH is bioavailable to the brain and under condition of inflammation exerts significant indirect effects on 2-AG levels. The biphenyl scaffold might serve as valuable source of dual CB2 receptor modulators and COX-2 SSIs as demonstrated by additional MH analogs that show similar effects. The combination of CB2 agonism and COX-2 SSI offers a yet unexplored polypharmacology with expected synergistic effects in neuroinflammatory diseases, thus providing a rationale for the diverse neuroprotective effects reported for MH in animal models.
Collapse
Affiliation(s)
- Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Maria Salomé Gachet
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Vanessa Petrucci
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Wolfgang Schuehly
- Institute of Zoology, Karl-Franzens-University Graz, Universitätsplatz 2, 8010, Graz, Austria.
| | - Roch-Philippe Charles
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bühlstrasse 28, CH-3012, Bern, Switzerland.
| |
Collapse
|
87
|
Dyall SC. Long-chain omega-3 fatty acids and the brain: a review of the independent and shared effects of EPA, DPA and DHA. Front Aging Neurosci 2015; 7:52. [PMID: 25954194 PMCID: PMC4404917 DOI: 10.3389/fnagi.2015.00052] [Citation(s) in RCA: 554] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/28/2015] [Indexed: 12/19/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) exhibit neuroprotective properties and represent a potential treatment for a variety of neurodegenerative and neurological disorders. However, traditionally there has been a lack of discrimination between the different omega-3 PUFAs and effects have been broadly accredited to the series as a whole. Evidence for unique effects of eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and more recently docosapentaenoic acid (DPA) is growing. For example, beneficial effects in mood disorders have more consistently been reported in clinical trials using EPA; whereas, with neurodegenerative conditions such as Alzheimer’s disease, the focus has been on DHA. DHA is quantitatively the most important omega-3 PUFA in the brain, and consequently the most studied, whereas the availability of high purity DPA preparations has been extremely limited until recently, limiting research into its effects. However, there is now a growing body of evidence indicating both independent and shared effects of EPA, DPA and DHA. The purpose of this review is to highlight how a detailed understanding of these effects is essential to improving understanding of their therapeutic potential. The review begins with an overview of omega-3 PUFA biochemistry and metabolism, with particular focus on the central nervous system (CNS), where DHA has unique and indispensable roles in neuronal membranes with levels preserved by multiple mechanisms. This is followed by a review of the different enzyme-derived anti-inflammatory mediators produced from EPA, DPA and DHA. Lastly, the relative protective effects of EPA, DPA and DHA in normal brain aging and the most common neurodegenerative disorders are discussed. With a greater understanding of the individual roles of EPA, DPA and DHA in brain health and repair it is hoped that appropriate dietary recommendations can be established and therapeutic interventions can be more targeted and refined.
Collapse
Affiliation(s)
- Simon C Dyall
- Faculty of Health and Social Sciences, Bournemouth University Bournemouth, UK
| |
Collapse
|
88
|
Renouard L, Billwiller F, Ogawa K, Clément O, Camargo N, Abdelkarim M, Gay N, Scoté-Blachon C, Touré R, Libourel PA, Ravassard P, Salvert D, Peyron C, Claustrat B, Léger L, Salin P, Malleret G, Fort P, Luppi PH. The supramammillary nucleus and the claustrum activate the cortex during REM sleep. SCIENCE ADVANCES 2015; 1:e1400177. [PMID: 26601158 PMCID: PMC4640625 DOI: 10.1126/sciadv.1400177] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/11/2015] [Indexed: 05/10/2023]
Abstract
Evidence in humans suggests that limbic cortices are more active during rapid eye movement (REM or paradoxical) sleep than during waking, a phenomenon fitting with the presence of vivid dreaming during this state. In that context, it seemed essential to determine which populations of cortical neurons are activated during REM sleep. Our aim in the present study is to fill this gap by combining gene expression analysis, functional neuroanatomy, and neurochemical lesions in rats. We find in rats that, during REM sleep hypersomnia compared to control and REM sleep deprivation, the dentate gyrus, claustrum, cortical amygdaloid nucleus, and medial entorhinal and retrosplenial cortices are the only cortical structures containing neurons with an increased expression of Bdnf, FOS, and ARC, known markers of activation and/or synaptic plasticity. Further, the dentate gyrus is the only cortical structure containing more FOS-labeled neurons during REM sleep hypersomnia than during waking. Combining FOS staining, retrograde labeling, and neurochemical lesion, we then provide evidence that FOS overexpression occurring in the cortex during REM sleep hypersomnia is due to projections from the supramammillary nucleus and the claustrum. Our results strongly suggest that only a subset of cortical and hippocampal neurons are activated and display plasticity during REM sleep by means of ascending projections from the claustrum and the supramammillary nucleus. Our results pave the way for future studies to identify the function of REM sleep with regard to dreaming and emotional memory processing.
Collapse
Affiliation(s)
- Leslie Renouard
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
- College of Medical Sciences, Washington State University, 412 E. Spokane Falls Boulevard, PBS230, Spokane, WA 99202, USA
| | - Francesca Billwiller
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Keiko Ogawa
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Olivier Clément
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Nutabi Camargo
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Mouaadh Abdelkarim
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Nadine Gay
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Céline Scoté-Blachon
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Rouguy Touré
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Paul-Antoine Libourel
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Pascal Ravassard
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Denise Salvert
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Christelle Peyron
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Bruno Claustrat
- Service de Radioanalyse, Centre de Médecine nucléaire, 59 Boulevard Pinel, 69677 Bron Cedex, France
| | - Lucienne Léger
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Paul Salin
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Gael Malleret
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Patrice Fort
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | - Pierre-Hervé Luppi
- UMR 5292 CNRS/U1028 INSERM, Centre de Recherche en Neurosciences de Lyon (CRNL), Team “Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil,” Université Claude Bernard Lyon 1, Faculté de Médecine RTH Laennec, 7 Rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| |
Collapse
|
89
|
Shiraki A, Saito F, Akane H, Akahori Y, Imatanaka N, Itahashi M, Yoshida T, Shibutani M. Gene expression profiling of the hippocampal dentate gyrus in an adult toxicity study captures a variety of neurodevelopmental dysfunctions in rat models of hypothyroidism. J Appl Toxicol 2015; 36:24-34. [DOI: 10.1002/jat.3140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/27/2015] [Accepted: 02/01/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Ayako Shiraki
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
- Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences; Gifu University; 1-1 Yanagido, Gifu-shi Gifu 501-1193 Japan
| | - Fumiyo Saito
- Chemicals Evaluation and Research Institute; Japan, 1-4-25 Koraku, Bunkyo-ku Tokyo 112-0004 Japan
| | - Hirotoshi Akane
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
| | - Yumi Akahori
- Chemicals Evaluation and Research Institute; Japan, 1-4-25 Koraku, Bunkyo-ku Tokyo 112-0004 Japan
| | - Nobuya Imatanaka
- Chemicals Evaluation and Research Institute; Japan, 1-4-25 Koraku, Bunkyo-ku Tokyo 112-0004 Japan
| | - Megu Itahashi
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
- Pathogenetic Veterinary Science, United Graduate School of Veterinary Sciences; Gifu University; 1-1 Yanagido, Gifu-shi Gifu 501-1193 Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology; Tokyo University of Agriculture and Technology; 3-5-8 Saiwai-cho, Fuchu-shi Tokyo 183-8509 Japan
| |
Collapse
|
90
|
Prakash A, Kalra J, Mani V, Ramasamy K, Majeed ABA. Pharmacological approaches for Alzheimer’s disease: neurotransmitter as drug targets. Expert Rev Neurother 2014; 15:53-71. [DOI: 10.1586/14737175.2015.988709] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
91
|
Comparative biochemical characterization of the monoacylglycerol lipase inhibitor KML29 in brain, spinal cord, liver, spleen, fat and muscle tissue. Neuropharmacology 2014; 91:148-56. [PMID: 25497453 DOI: 10.1016/j.neuropharm.2014.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/08/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022]
Abstract
Monoacylglycerol lipase (MAGL) is part of the endocannabinoid and the prostaglandin signaling system. MAGL degrades the endocannabinoid 2-arachidonoylglycerol (2-AG) into glycerol and arachidonic acid. MAGL-induced arachidonic acid is the primary source for prostaglandin synthesis in the brain. 2-AG mainly induces neuroprotective and anti-inflammatory effects, whereas prostaglandins are related to pro-inflammatory effects inducing neurotoxicity. Therefore, inhibition of MAGL represents a promising target for neurological diseases characterized by inflammation. However, as 2-AG is an agonist for the cannabinoid receptor 1 (CB1), inhibition of MAGL might be associated with unwanted cannabimimetic effects. Here, we show that oral administration of KML29, a highly selective inhibitor of MAGL, induced large and dose-dependent changes in 2-AG levels in vivo in brain and spinal cord of mice. Of note, MAGL inhibition by KML29 induced a decrease in prostaglandin levels in brain and most peripheral tissues but not in the spinal cord. MAGL expression was highest in fat, liver and brain, whereas the cytosolic phospholipase A2 (cPLA2), a further enzyme responsible for arachidonic acid production, was highly expressed in spinal cord, muscle and spleen. In addition, high doses (10 mg/kg) of KML29 induced some cannabimimetic effects in vivo in the tetrad test, including hypothermia, analgesia and hypomotility without induction of cataleptic behavior. In summary, inhibition of MAGL by KML29 represents a promising strategy for targeting the cannabinoid and prostaglandin system of the brain with only a moderate induction of cannabimimetic effects.
Collapse
|
92
|
García del Caño G, Aretxabala X, González-Burguera I, Montaña M, López de Jesús M, Barrondo S, Barrio RJ, Sampedro C, Goicolea MA, Sallés J. Nuclear diacylglycerol lipase-α in rat brain cortical neurons: evidence of 2-arachidonoylglycerol production in concert with phospholipase C-β activity. J Neurochem 2014; 132:489-503. [PMID: 25308538 DOI: 10.1111/jnc.12963] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/22/2014] [Accepted: 10/07/2014] [Indexed: 01/12/2023]
Abstract
In this report, we describe the localization of diacylglycerol lipase-α (DAGLα) in nuclei from adult cortical neurons, as assessed by double-immunofluorescence staining of rat brain cortical sections and purified intact nuclei and by western blot analysis of subnuclear fractions. Double-labeling assays using the anti-DAGLα antibody and NeuN combined with Hoechst staining showed that only nuclei of neuronal origin were DAGLα positive. At high resolution, DAGLα-signal displayed a punctate pattern in nuclear subdomains poor in Hoechst's chromatin and lamin B1 staining. In contrast, SC-35- and NeuN-signals (markers of the nuclear speckles) showed a high overlap with DAGLα within specific subdomains of the nuclear matrix. Among the members of the phospholipase C-β (PLCβ) family, PLCβ1, PLCβ2, and PLCβ4 exhibited the same distribution with respect to chromatin, lamin B1, SC-35, and NeuN as that described for DAGLα. Furthermore, by quantifying the basal levels of 2-arachidonoylglycerol (2-AG) by liquid chromatography and mass spectrometry (LC-MS), and by characterizing the pharmacology of its accumulation, we describe the presence of a mechanism for 2-AG production, and its PLCβ/DAGLα-dependent biosynthesis in isolated nuclei. These results extend our knowledge about subcellular distribution of neuronal DAGLα, providing biochemical grounds to hypothesize a role for 2-AG locally produced within the neuronal nucleus.
Collapse
Affiliation(s)
- Gontzal García del Caño
- Departamento de Neurociencias, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Xabier Aretxabala
- Departamento de Neurociencias, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Imanol González-Burguera
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Mario Montaña
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Maider López de Jesús
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Sergio Barrondo
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Ramón J Barrio
- Departamento de Química Analítica, Facultad de Farmacia, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Carmen Sampedro
- Servicio General de Análisis, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - M Arantzazu Goicolea
- Departamento de Química Analítica, Facultad de Farmacia, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| | - Joan Sallés
- Departamento de Farmacología, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 01006 Vitoria-Gasteiz (Araba), Spain
| |
Collapse
|
93
|
Primiani CT, Ryan VH, Rao JS, Cam MC, Ahn K, Modi HR, Rapoport SI. Coordinated gene expression of neuroinflammatory and cell signaling markers in dorsolateral prefrontal cortex during human brain development and aging. PLoS One 2014; 9:e110972. [PMID: 25329999 PMCID: PMC4203852 DOI: 10.1371/journal.pone.0110972] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/17/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Age changes in expression of inflammatory, synaptic, and neurotrophic genes are not well characterized during human brain development and senescence. Knowing these changes may elucidate structural, metabolic, and functional brain processes over the lifespan, as well vulnerability to neurodevelopmental or neurodegenerative diseases. HYPOTHESIS Expression levels of inflammatory, synaptic, and neurotrophic genes in the human brain are coordinated over the lifespan and underlie changes in phenotypic networks or cascades. METHODS We used a large-scale microarray dataset from human prefrontal cortex, BrainCloud, to quantify age changes over the lifespan, divided into Development (0 to 21 years, 87 brains) and Aging (22 to 78 years, 144 brains) intervals, in transcription levels of 39 genes. RESULTS Gene expression levels followed different trajectories over the lifespan. Many changes were intercorrelated within three similar groups or clusters of genes during both Development and Aging, despite different roles of the gene products in the two intervals. During Development, changes were related to reported neuronal loss, dendritic growth and pruning, and microglial events; TLR4, IL1R1, NFKB1, MOBP, PLA2G4A, and PTGS2 expression increased in the first years of life, while expression of synaptic genes GAP43 and DBN1 decreased, before reaching plateaus. During Aging, expression was upregulated for potentially pro-inflammatory genes such as NFKB1, TRAF6, TLR4, IL1R1, TSPO, and GFAP, but downregulated for neurotrophic and synaptic integrity genes such as BDNF, NGF, PDGFA, SYN, and DBN1. CONCLUSIONS Coordinated changes in gene transcription cascades underlie changes in synaptic, neurotrophic, and inflammatory phenotypic networks during brain Development and Aging. Early postnatal expression changes relate to neuronal, glial, and myelin growth and synaptic pruning events, while late Aging is associated with pro-inflammatory and synaptic loss changes. Thus, comparable transcriptional regulatory networks that operate throughout the lifespan underlie different phenotypic processes during Aging compared to Development.
Collapse
Affiliation(s)
- Christopher T. Primiani
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Veronica H. Ryan
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jagadeesh S. Rao
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Margaret C. Cam
- Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kwangmi Ahn
- Child Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hiren R. Modi
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stanley I. Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
94
|
Trandafir CC, Pouliot WA, Dudek FE, Ekstrand JJ. Co-administration of subtherapeutic diazepam enhances neuroprotective effect of COX-2 inhibitor, NS-398, after lithium pilocarpine-induced status epilepticus. Neuroscience 2014; 284:601-610. [PMID: 25453777 DOI: 10.1016/j.neuroscience.2014.10.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 12/19/2022]
Abstract
RATIONALE Seizures during status epilepticus (SE) cause neuronal death and induce cyclooxygenase-2 (COX-2). Pilocarpine-induced SE was used to determine if COX-2 inhibition with NS-398, when administered alone or with diazepam, decreases the duration and/or intensity of SE and/or reduces neuronal injury in the rat hippocampus. METHODS Electroencephalogram (EEG) electrodes were implanted in male Sprague-Dawley rats. SE was induced with lithium-pilocarpine, and continuous EEG and video monitoring were performed for 24 h. Rats were divided into four groups (n=8-14 rats/group) and received NS-398, diazepam, NS-398 and diazepam, or vehicle 30 min after the first motor seizure. Six hours later, NS-398 injection was repeated in the NS-398 and in the NS-398+diazepam groups. The duration of SE (continuous spiking) and the EEG power in the γ-band were analyzed. FluoroJade B staining in the dorsal hippocampus at 24h after SE was analyzed semi-quantitatively in the CA1, CA3 and hilus. RESULTS The duration and intensity of electrographic SE was not significantly different across the four groups. In rats treated with NS-398 alone, compared to vehicle-treated rats, neuronal damage was significantly lower compared to vehicle-treated rats in the CA3 (27%) and hilus (27%), but neuroprotection was not detected in the CA1. When NS-398 was administered with diazepam, decreased neuronal damage was further obtained in all areas investigated (CA1: 61%, CA3: 63%, hilus: 60%). CONCLUSIONS NS-398, when administered 30 min after the onset of SE with a repeat dose at 6h, decreased neuronal damage in the hippocampus. Administration of diazepam with NS-398 potentiates the neuroprotective effect of the COX-2 inhibitor. These neuroprotective effects occurred with no detectable effect on electrographic SE.
Collapse
Affiliation(s)
- C C Trandafir
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, United States
| | - W A Pouliot
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, United States
| | - F E Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, United States
| | - J J Ekstrand
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, United States.
| |
Collapse
|
95
|
Wei J, Du K, Cai Q, Ma L, Jiao Z, Tan J, Xu Z, Li J, Luo W, Chen J, Gao J, Zhang D, Huang C. Lead induces COX-2 expression in glial cells in a NFAT-dependent, AP-1/NFκB-independent manner. Toxicology 2014; 325:67-73. [PMID: 25193092 DOI: 10.1016/j.tox.2014.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/22/2022]
Abstract
Epidemiologic studies have provided solid evidence for the neurotoxic effect of lead for decades of years. In view of the fact that children are more vulnerable to the neurotoxicity of lead, lead exposure has been an urgent public health concern. The modes of action of lead neurotoxic effects include disturbance of neurotransmitter storage and release, damage of mitochondria, as well as induction of apoptosis in neurons, cerebrovascular endothelial cells, astroglia and oligodendroglia. Our studies here, from a novel point of view, demonstrates that lead specifically caused induction of COX-2, a well known inflammatory mediator in neurons and glia cells. Furthermore, we revealed that COX-2 was induced by lead in a transcription-dependent manner, which relayed on transcription factor NFAT, rather than AP-1 and NFκB, in glial cells. Considering the important functions of COX-2 in mediation of inflammation reaction and oxidative stress, our studies here provide a mechanistic insight into the understanding of lead-associated inflammatory neurotoxicity effect via activation of pro-inflammatory NFAT3/COX-2 axis.
Collapse
Affiliation(s)
- Jinlong Wei
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Kejun Du
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA; Department of Occupational and Environmental Health and Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Qinzhen Cai
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lisha Ma
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhenzhen Jiao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jinrong Tan
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhou Xu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Wenjin Luo
- Department of Occupational and Environmental Health and Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Jingyuan Chen
- Department of Occupational and Environmental Health and Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| |
Collapse
|
96
|
Hermanson DJ, Gamble-George JC, Marnett LJ, Patel S. Substrate-selective COX-2 inhibition as a novel strategy for therapeutic endocannabinoid augmentation. Trends Pharmacol Sci 2014; 35:358-67. [PMID: 24845457 PMCID: PMC4074568 DOI: 10.1016/j.tips.2014.04.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 12/20/2022]
Abstract
Pharmacologic augmentation of endogenous cannabinoid (eCB) signaling is an emerging therapeutic approach for the treatment of a broad range of pathophysiological conditions. Thus far, pharmacological approaches have focused on inhibition of the canonical eCB inactivation pathways - fatty acid amide hydrolase (FAAH) for anandamide and monoacylglycerol lipase (MAGL) for 2-arachidonoylglycerol. We review here the experimental evidence that cyclooxygenase-2 (COX-2)-mediated eCB oxygenation represents a third mechanism for terminating eCB action at cannabinoid receptors. We describe the development, molecular mechanisms, and in vivo validation of 'substrate-selective' COX-2 inhibitors (SSCIs) that prevent eCB inactivation by COX-2 without affecting prostaglandin (PG) generation from arachidonic acid (AA). Lastly, we review recent data on the potential therapeutic applications of SSCIs with a focus on neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daniel J Hermanson
- A.B. Hancock Jr Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology Center in Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Joyonna C Gamble-George
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lawrence J Marnett
- A.B. Hancock Jr Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology Center in Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Sachin Patel
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
97
|
Coordination of gene expression of arachidonic and docosahexaenoic acid cascade enzymes during human brain development and aging. PLoS One 2014; 9:e100858. [PMID: 24963629 PMCID: PMC4070994 DOI: 10.1371/journal.pone.0100858] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 05/30/2014] [Indexed: 12/19/2022] Open
Abstract
Background The polyunsaturated arachidonic and docosahexaenoic acids (AA and DHA) participate in cell membrane synthesis during neurodevelopment, neuroplasticity, and neurotransmission throughout life. Each is metabolized via coupled enzymatic reactions within separate but interacting metabolic cascades. Hypothesis AA and DHA pathway genes are coordinately expressed and underlie cascade interactions during human brain development and aging. Methods The BrainCloud database for human non-pathological prefrontal cortex gene expression was used to quantify postnatal age changes in mRNA expression of 34 genes involved in AA and DHA metabolism. Results Expression patterns were split into Development (0 to 20 years) and Aging (21 to 78 years) intervals. Expression of genes for cytosolic phospholipases A2 (cPLA2), cyclooxygenases (COX)-1 and -2, and other AA cascade enzymes, correlated closely with age during Development, less so during Aging. Expression of DHA cascade enzymes was less inter-correlated in each period, but often changed in the opposite direction to expression of AA cascade genes. Except for the PLA2G4A (cPLA2 IVA) and PTGS2 (COX-2) genes at 1q25, highly inter-correlated genes were at distant chromosomal loci. Conclusions Coordinated age-related gene expression during the brain Development and Aging intervals likely underlies coupled changes in enzymes of the AA and DHA cascades and largely occur through distant transcriptional regulation. Healthy brain aging does not show upregulation of PLA2G4 or PTGS2 expression, which was found in Alzheimer's disease.
Collapse
|
98
|
Rapoport SI. Lithium and the other mood stabilizers effective in bipolar disorder target the rat brain arachidonic acid cascade. ACS Chem Neurosci 2014; 5:459-67. [PMID: 24786695 DOI: 10.1021/cn500058v] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
This Review evaluates the arachidonic acid (AA, 20:4n-6) cascade hypothesis for the actions of lithium and other FDA-approved mood stabilizers in bipolar disorder (BD). The hypothesis is based on evidence in unanesthetized rats that chronically administered lithium, carbamazepine, valproate, or lamotrigine each downregulated brain AA metabolism, and it is consistent with reported upregulated AA cascade markers in post-mortem BD brain. In the rats, each mood stabilizer reduced AA turnover in brain phospholipids, cyclooxygenase-2 expression, and prostaglandin E2 concentration. Lithium and carbamazepine also reduced expression of cytosolic phospholipase A2 (cPLA2) IVA, which releases AA from membrane phospholipids, whereas valproate uncompetitively inhibited in vitro acyl-CoA synthetase-4, which recycles AA into phospholipid. Topiramate and gabapentin, proven ineffective in BD, changed rat brain AA metabolism minimally. On the other hand, the atypical antipsychotics olanzapine and clozapine, which show efficacy in BD, decreased rat brain AA metabolism by reducing plasma AA availability. Each of the four approved mood stabilizers also dampened brain AA signaling during glutamatergic NMDA and dopaminergic D2 receptor activation, while lithium enhanced the signal during cholinergic muscarinic receptor activation. In BD patients, such signaling effects might normalize the neurotransmission imbalance proposed to cause disease symptoms. Additionally, the antidepressants fluoxetine and imipramine, which tend to switch BD depression to mania, each increased AA turnover and cPLA2 IVA expression in rat brain, suggesting that brain AA metabolism is higher in BD mania than depression. The AA hypothesis for mood stabilizer action is consistent with reports that low-dose aspirin reduced morbidity in patients taking lithium, and that high n-3 and/or low n-6 polyunsaturated fatty acid diets, which in rats reduce brain AA metabolism, were effective in BD and migraine patients.
Collapse
Affiliation(s)
- Stanley I. Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
99
|
An Y, Belevych N, Wang Y, Zhang H, Nasse JS, Herschman H, Chen Q, Tarr A, Liu X, Quan N. Prostacyclin mediates endothelial COX-2-dependent neuroprotective effects during excitotoxic brain injury. J Inflamm Res 2014; 7:57-67. [PMID: 24971026 PMCID: PMC4070856 DOI: 10.2147/jir.s63205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In a previous study, we found that intracerebral administration of excitotoxin (RS)-(tetrazole-5yl) glycine caused increased neural damage in the brain in an endothelial COX-2 deleted mouse line (Tie2Cre COX-2(flox/flox) ). In this study, we investigated whether prostacyclin might mediate this endothelial COX-2-dependent neuroprotection. Administration of excitotoxin into the striatum induced the production of prostacyclin (PGI2) in wild type, but not in endothelial COX-2 deleted mice. Inhibition of PGI2 synthase exacerbated brain lesions induced by the excitotoxin in wild type, but not in endothelial COX-2 deleted mice. Administration of a PGI2 agonist reduced neural damage in both wild type and endothelial COX-2 deleted mice. Increased PGI2 synthase expression was found in infiltrating neutrophils. In an ex vivo assay, PGI2 reduced the excitotoxin-induced calcium influx into neurons, suggesting a cellular mechanism for PGI2 mediated neuroprotection. These results reveal that PGI2 mediates endothelial COX-2 dependent neuroprotection.
Collapse
Affiliation(s)
- Ying An
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| | - Natalya Belevych
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| | - Yufen Wang
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| | - Hao Zhang
- Institute for Behavior Medicine Research, Columbus, OH, USA
| | - Jason S Nasse
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA
| | - Harvey Herschman
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Qun Chen
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| | - Andrew Tarr
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| | - Xiaoyu Liu
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| | - Ning Quan
- Institute for Behavior Medicine Research, Columbus, OH, USA ; Department of Oral Biology, College of Dentistry, Columbus, OH, USA
| |
Collapse
|
100
|
The structure of the geriatric depressed brain and response to electroconvulsive therapy. Psychiatry Res 2014; 222:1-9. [PMID: 24686000 DOI: 10.1016/j.pscychresns.2014.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 02/08/2023]
Abstract
Electroconvulsive therapy (ECT) is the treatment of choice in severe geriatric depression. High remission rates may be influenced by specific brain morphology characteristic of geriatric depression. Our objective was to identify the relationship between brain structure, symptom profile, and ECT response. In a naturalistic cohort of 55 patients with a major depressive disorder, structural magnetic resonance imaging (MRI) was performed before ECT. Voxel-based morphometry was applied to determine regional differences in gray matter (GM) volume between patients and 23 matched healthy controls. Depressed patients with psychotic symptoms showed significantly higher remission rates and smaller regional GM volume of the left inferior frontal gyrus (IFG). Patients with late onset depression showed smaller regional GM volume of the bilateral lateral temporal cortex. Larger size of response in the whole patient group was related to smaller pretreatment regional GM volume of the right lateral temporal cortex, whereas faster speed of response was related to smaller pretreatment regional GM volume of the right IFG. ECT is most effective in depressed patients with psychotic symptoms. In this study the presence of psychotic symptoms was related to pretreatment smaller GM volume of the left IFG and bilateral temporal cortex. Smaller volume of the IFG pretreatment was related to faster treatment response, and smaller volume of the right lateral temporal cortex pretreatment was related to larger response to ECT. These results are possibly explained by the connectivity between these brain regions and an interconnected network that is particularly activated by the ECT-induced seizures.
Collapse
|