1
|
Aruna K, Pal S, Khanna A, Bhattacharyya S. Postsynaptic Density Proteins and Their Role in the Trafficking of Group I Metabotropic Glutamate Receptors. J Membr Biol 2024; 257:257-268. [PMID: 39369356 DOI: 10.1007/s00232-024-00326-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system that regulates multiple different forms of synaptic plasticity, including learning and memory. Glutamate transduces its signal by activating ionotropic glutamate receptors and metabotropic glutamate receptors (mGluRs). Group I mGluRs belong to the G protein-coupled receptor (GPCR) family. Regulation of cell surface expression and trafficking of the glutamate receptors represents an important mechanism that assures proper transmission of information at the synapses. There is growing evidence implicating dysregulated glutamate receptor trafficking in the pathophysiology of several neuropsychiatric disorders. The postsynaptic density (PSD) region consists of many specialized proteins which are assembled beneath the postsynaptic membrane of dendritic spines. Many of these proteins interact with group I mGluRs and have essential roles in group I mGluR-mediated synaptic function and plasticity. This review provides up-to-date information on the molecular determinants regulating cell surface expression and trafficking of group I mGluRs and discusses the role of few of these PSD proteins in these processes. As substantial evidences link mGluR dysfunction and maladaptive functioning of many PSD proteins to the pathophysiology of various neuropsychiatric disorders, understanding the role of the PSD proteins in group I mGluR trafficking may provide opportunities for the development of novel therapeutics in multiple neuropsychiatric disorders.
Collapse
Affiliation(s)
- K Aruna
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, Punjab, 140306, India
| | - Subhajit Pal
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, Punjab, 140306, India
| | - Ankita Khanna
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, Punjab, 140306, India
| | - Samarjit Bhattacharyya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge city, Sector - 81, SAS Nagar, Punjab, 140306, India.
| |
Collapse
|
2
|
Dong LG, An MQ, Gu HY, Zhang LG, Zhang JB, Li CJ, Mao CJ, Wang F, Liu CF. PACAP/PAC1-R activation contributes to hyperalgesia in 6-OHDA-induced Parkinson's disease model rats via promoting excitatory synaptic transmission of spinal dorsal horn neurons. Acta Pharmacol Sin 2023; 44:2418-2431. [PMID: 37563446 PMCID: PMC10692161 DOI: 10.1038/s41401-023-01141-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023]
Abstract
Pain is a common annoying non-motor symptom in Parkinson's disease (PD) that causes distress to patients. Treatment for PD pain remains a big challenge, as its underlying mechanisms are elusive. Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor PAC1-R play important roles in regulating a variety of pathophysiological processes. In this study, we investigated whether PACAP/PAC1-R signaling was involved in the mechanisms of PD pain. 6-hydroxydopamine (6-OHDA)-induced PD model was established in rats. Behavioral tests, electrophysiological and Western blotting analysis were conducted 3 weeks later. We found that 6-OHDA rats had significantly lower mechanical paw withdrawal 50% threshold in von Frey filament test and shorter tail flick latency, while mRNA levels of Pacap and Adcyap1r1 (gene encoding PAC1-R) in the spinal dorsal horn were significantly upregulated. Whole-cell recordings from coronal spinal cord slices at L4-L6 revealed that the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in dorsal horn neurons was significantly increased, which was reversed by application of a PAC1-R antagonist PACAP 6-38 (250 nM). Furthermore, we demonstrated that intrathecal microinjection of PACAP 6-38 (0.125, 0.5, 2 μg) dose-dependently ameliorated the mechanical and thermal hyperalgesia in 6-OHDA rats. Inhibition of PACAP/PAC1-R signaling significantly suppressed the activation of Ca2+/calmodulin-dependent protein kinase II and extracellular signal-regulated kinase (ERK) in spinal dorsal horn of 6-OHDA rats. Microinjection of pAAV-Adcyap1r1 into L4-L6 spinal dorsal horn alleviated hyperalgesia in 6-OHDA rats. Intrathecal microinjection of ERK antagonist PD98059 (10 μg) significantly alleviated hyperalgesia in 6-OHDA rats associated with the inhibition of sEPSCs in dorsal horn neurons. In addition, we found that serum PACAP-38 concentration was significantly increased in PD patients with pain, and positively correlated with numerical rating scale score. In conclusion, activation of PACAP/PAC1-R induces the development of PD pain and targeting PACAP/PAC1-R is an alternative strategy for treating PD pain.
Collapse
Affiliation(s)
- Li-Guo Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Meng-Qi An
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Han-Ying Gu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Li-Ge Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Jin-Bao Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Cheng-Jie Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China.
| |
Collapse
|
3
|
de Bartolomeis A, Ciccarelli M, De Simone G, Mazza B, Barone A, Vellucci L. Canonical and Non-Canonical Antipsychotics' Dopamine-Related Mechanisms of Present and Next Generation Molecules: A Systematic Review on Translational Highlights for Treatment Response and Treatment-Resistant Schizophrenia. Int J Mol Sci 2023; 24:ijms24065945. [PMID: 36983018 PMCID: PMC10051989 DOI: 10.3390/ijms24065945] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Schizophrenia is a severe psychiatric illness affecting almost 25 million people worldwide and is conceptualized as a disorder of synaptic plasticity and brain connectivity. Antipsychotics are the primary pharmacological treatment after more than sixty years after their introduction in therapy. Two findings hold true for all presently available antipsychotics. First, all antipsychotics occupy the dopamine D2 receptor (D2R) as an antagonist or partial agonist, even if with different affinity; second, D2R occupancy is the necessary and probably the sufficient mechanism for antipsychotic effect despite the complexity of antipsychotics' receptor profile. D2R occupancy is followed by coincident or divergent intracellular mechanisms, implying the contribution of cAMP regulation, β-arrestin recruitment, and phospholipase A activation, to quote some of the mechanisms considered canonical. However, in recent years, novel mechanisms related to dopamine function beyond or together with D2R occupancy have emerged. Among these potentially non-canonical mechanisms, the role of Na2+ channels at the dopamine at the presynaptic site, dopamine transporter (DAT) involvement as the main regulator of dopamine concentration at synaptic clefts, and the putative role of antipsychotics as chaperones for intracellular D2R sequestration, should be included. These mechanisms expand the fundamental role of dopamine in schizophrenia therapy and may have relevance to considering putatively new strategies for treatment-resistant schizophrenia (TRS), an extremely severe condition epidemiologically relevant and affecting almost 30% of schizophrenia patients. Here, we performed a critical evaluation of the role of antipsychotics in synaptic plasticity, focusing on their canonical and non-canonical mechanisms of action relevant to the treatment of schizophrenia and their subsequent implication for the pathophysiology and potential therapy of TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
4
|
Galineau L, Arlicot N, Dupont AC, Briend F, Houy-Durand E, Tauber C, Gomot M, Gissot V, Barantin L, Lefevre A, Vercouillie J, Roussel C, Roux S, Nadal L, Mavel S, Laumonnier F, Belzung C, Chalon S, Emond P, Santiago-Ribeiro MJ, Bonnet-Brilhault F. Glutamatergic synapse in autism: a complex story for a complex disorder. Mol Psychiatry 2023; 28:801-809. [PMID: 36434055 DOI: 10.1038/s41380-022-01860-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/12/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose pathophysiological mechanisms are still unclear. Hypotheses suggest a role for glutamate dysfunctions in ASD development, but clinical studies investigating brain and peripheral glutamate levels showed heterogenous results leading to hypo- and hyper-glutamatergic hypotheses of ASD. Recently, studies proposed the implication of elevated mGluR5 densities in brain areas in the pathophysiology of ASD. Thus, our objective was to characterize glutamate dysfunctions in adult subjects with ASD by quantifying (1) glutamate levels in the cingulate cortex and periphery using proton magnetic resonance spectroscopy and metabolomics, and (2) mGluR5 brain density in this population and in a validated animal model of ASD (prenatal exposure to valproate) at developmental stages corresponding to childhood and adolescence in humans using positron emission tomography. No modifications in cingulate Glu levels were observed between individuals with ASD and controls further supporting the difficulty to evaluate modifications in excitatory transmission using spectroscopy in this population, and the complexity of its glutamate-related changes. Our imaging results showed an overall increased density in mGluR5 in adults with ASD, that was only observed mostly subcortically in adolescent male rats prenatally exposed to valproic acid, and not detected in the stage corresponding to childhood in the same animals. This suggest that clinical changes in mGluR5 density could reflect the adaptation of the glutamatergic dysfunctions occurring earlier rather than being key to the pathophysiology of ASD.
Collapse
Affiliation(s)
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Unité de Radiopharmacie, CHRU de Tours, Tours, France
| | - Anne-Claire Dupont
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Unité de Radiopharmacie, CHRU de Tours, Tours, France.,Service de Médecine Nucléaire, CHRU de Tours, Tours, France
| | - Frederic Briend
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | - Emmanuelle Houy-Durand
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | - Clovis Tauber
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Marie Gomot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | | | | | - Antoine Lefevre
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | | | - Sylvie Roux
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | - Lydie Nadal
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvie Mavel
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | | | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Maria-Joao Santiago-Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Service de Médecine Nucléaire, CHRU de Tours, Tours, France
| | - Frédérique Bonnet-Brilhault
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France. .,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France.
| |
Collapse
|
5
|
Genetic and genomic signatures in ethanol withdrawal seizure-prone and seizure-resistant mice implicate genes involved in epilepsy and neuronal excitability. Mol Psychiatry 2022; 27:4611-4623. [PMID: 36198764 DOI: 10.1038/s41380-022-01799-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Alcohol withdrawal is a clinically important consequence and potential driver of Alcohol Use Disorder. However, susceptibility to withdrawal symptoms, ranging from craving and anxiety to seizures and delirium, varies greatly. Selectively bred Withdrawal Seizure-Prone (WSP) and Seizure-Resistant (WSR) mice are an animal model of differential susceptibility to withdrawal and phenotypes with which withdrawal severity correlates. To identify innate drivers of alcohol withdrawal severity, we performed a multi-omic study of the WSP and WSR lines and F2 mice derived from them, using genomic, genetic, and transcriptomic analyses. Genes implicated in seizures and epilepsy were over-represented among those that segregated between WSP and WSR mice and that displayed differential expression in F2 mice high and low in withdrawal. Quantitative trait locus (QTL) analysis of ethanol withdrawal convulsions identified several genome-wide significant loci and pointed to genes that modulate potassium channel function and neural excitability. Perturbations of expression of genes involved in synaptic transmission, including GABAergic and glutamatergic genes, were prominent in prefrontal cortex transcriptome. Expression QTL (eQTL) analysis fine mapped genes within the peak ethanol withdrawal QTL regions. Genetic association analysis in human subjects provided converging evidence for the involvement of those genes in severity of alcohol withdrawal and dependence. Our results reveal a polygenic network and neural signaling pathways contributing to ethanol withdrawal seizures and related phenotypes that overlap with genes modulating epilepsy and neuronal excitability.
Collapse
|
6
|
Huang XJ, Su GJ, Wu CW, Sha XS, Zou JF, Liu XS, Li M, He Y. Knockdown of rno_circRNA_009194 Improves Outcomes in Traumatic Brain Injury Rats through Inhibiting Voltage-Gated Sodium Channel Nav1.3. J Neurotrauma 2021; 39:196-210. [PMID: 34726508 DOI: 10.1089/neu.2020.7520] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Excessive activation of voltage-gated sodium channel Nav1.3 has been recently reported in secondary traumatic brain injury (TBI). However, the molecular mechanisms underlying regulating voltage-gated sodium channel (Nav1.3) have not been well understood. The present study used a TBI rat model induced by a fluid percussion device and performed a circular RNA (circRNA) microarray (n = 3) to profile the altered circRNAs in the hippocampus after TBI. After polymerase chain reaction (PCR) validation, certain circRNAs were selected to investigate the function and mechanism in regulating Nav1.3 in the TBI rat model by intracerebroventricular injection with lentivirus. The neurological outcome was evaluated by Morris water maze test, modified Neurological Severity Score (mNSS), brain water content measurement, and hematoxylin and eosin staining. The related molecular mechanisms were explored with PCR, Western blotting, luciferase reporter, chromatin immunoprecipitation assay, and electrophoretic mobility shift assay (EMSA). A total of 347 circRNAs were observed to be differentially expressed (fold change [FC] ≥ 1.2 and p < 0.05) after TBI, including 234 up-regulated and 113 down-regulated circRNAs. Among 10 validated circRNAs, we selected circRNA_009194 with the maximized up-regulated fold change (n = 5, FC = 4.45, p < 0.001) for the in vivo functional experiments. Down-regulation of circRNA_009194 resulted in a 27.5% reduced mNSS in rat brain (n = 6, p < 0.01) after TBI and regulated the expression levels of miR-145-3p, Sp1, and Nav1.3, which was reversed by sh-miR-145-3p or Sp1/Nav1.3 overexpression (n = 5, p < 0.05). Mechanistically, circRNA_009194 might act as a sponge for miR-145-3p to regulate Sp1-mediated Nav1.3. This study demonstrated that circRNA_009194 knockdown could improve neurological outcomes in TBI in vivo by inhibiting Nav1.3, directly or indirectly.
Collapse
Affiliation(s)
- Xian-Jian Huang
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Gao-Jian Su
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Chu-Wei Wu
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Xiao-Song Sha
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jun-Feng Zou
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Xian-Sheng Liu
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Min Li
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Yun He
- Department of Intensive Care Unit, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Shi G, Yin C, Fan Z, Xing L, Mostovoy Y, Kwok PY, Ashbrook LH, Krystal AD, Ptáček LJ, Fu YH. Mutations in Metabotropic Glutamate Receptor 1 Contribute to Natural Short Sleep Trait. Curr Biol 2020; 31:13-24.e4. [PMID: 33065013 DOI: 10.1016/j.cub.2020.09.071] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 01/03/2023]
Abstract
Sufficient and efficient sleep is crucial for our health. Natural short sleepers can sleep significantly shorter than the average population without a desire for more sleep and without any obvious negative health consequences. In searching for genetic variants underlying the short sleep trait, we found two different mutations in the same gene (metabotropic glutamate receptor 1) from two independent natural short sleep families. In vitro, both of the mutations exhibited loss of function in receptor-mediated signaling. In vivo, the mice carrying the individual mutations both demonstrated short sleep behavior. In brain slices, both of the mutations changed the electrical properties and increased excitatory synaptic transmission. These results highlight the important role of metabotropic glutamate receptor 1 in modulating sleep duration.
Collapse
Affiliation(s)
- Guangsen Shi
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chen Yin
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Zenghua Fan
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lijuan Xing
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yulia Mostovoy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Pui-Yan Kwok
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Liza H Ashbrook
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrew D Krystal
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Louis J Ptáček
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Ying-Hui Fu
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Glutamatergic postsynaptic density in early life stress programming: Topographic gene expression of mGlu5 receptors and Homer proteins. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109725. [PMID: 31404590 DOI: 10.1016/j.pnpbp.2019.109725] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/24/2019] [Accepted: 07/28/2019] [Indexed: 11/24/2022]
Abstract
Type-5 metabotropic glutamate receptors (mGlu5) have been implicated in the mechanism of resilience to stress. They form part of the postsynaptic density (PSD), a thickening of the glutamatergic synapse that acts as a multimodal hub for multiple cellular signaling. Perinatal stress in rats triggers alterations that make adult offspring less resilient to stress. In the present study, we examined the expression of gene encoding the mGlu5 (Grm5), as well as those encoding the short and long isoforms of Homer proteins in different brain regions of the offspring of dams exposed to repeated episodes of restraint stress during pregnancy ("perinatally stressed" or PRS offspring). To this end, we investigated unconditioned behavioral response using the light/dark box test, as well as the expression of PSD genes (Homer1a, Homer1b, and Grm5), in the medial prefrontal cortex, cortex, caudate-putamen, amygdala, and dorsal hippocampus. PRS rats spent significantly less time in the light area than the control group. In the amygdala, Homer1a mRNA levels were significantly increased in PRS rats, whereas Homer1b and Grm5 mRNA levels were reduced. In contrast, the transcript encoding for Homer1a was significantly reduced in the medial prefrontal cortex, caudate-putamen, and dorsal hippocampus of PRS rats. We also evaluated the relative ratio between Homer1a and Homer1b/Grm5 expression, finding a significant shift toward the expression of Homer1a in the amygdala and toward Homer1b/Grm5 in the other brain regions. These topographic patterns of Homer1a, Homer1b, and mGlu5 gene expression were significantly correlated with risk-taking behavior measured in the light/dark box test. Remarkably, in the amygdala and in other brain regions, Homer1b and Grm5 expression showed positive correlation with time spent in the light box, whereas Homer1a in the amygdala showed a negative correlation with risk-taking behavior, in contrast with all other brain regions analyzed, wherein these correlations were positive. These results suggest that perinatal stress programs the developmental expression of PSD molecules involved in mGlu5 signaling in discrete brain regions, with a predominant role for the amygdala.
Collapse
|
9
|
Serchov T, Schwarz I, Theiss A, Sun L, Holz A, Döbrössy MD, Schwarz MK, Normann C, Biber K, van Calker D. Enhanced adenosine A 1 receptor and Homer1a expression in hippocampus modulates the resilience to stress-induced depression-like behavior. Neuropharmacology 2019; 162:107834. [PMID: 31682853 DOI: 10.1016/j.neuropharm.2019.107834] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 11/30/2022]
Abstract
Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.
Collapse
Affiliation(s)
- Tsvetan Serchov
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| | - Inna Schwarz
- Functional Neuroconnectomics Group, Department of Experimental Epileptology and Cognition Research, Life and Brain Centre, University of Bonn, Medical School, 53105, Bonn, Germany
| | - Alice Theiss
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| | - Lu Sun
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713, AV Groningen, the Netherlands
| | - Amrei Holz
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Mate D Döbrössy
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
| | - Martin K Schwarz
- Functional Neuroconnectomics Group, Department of Experimental Epileptology and Cognition Research, Life and Brain Centre, University of Bonn, Medical School, 53105, Bonn, Germany
| | - Claus Normann
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| | - Knut Biber
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713, AV Groningen, the Netherlands
| | - Dietrich van Calker
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| |
Collapse
|
10
|
Fonin AV, Darling AL, Kuznetsova IM, Turoverov KK, Uversky VN. Multi-functionality of proteins involved in GPCR and G protein signaling: making sense of structure-function continuum with intrinsic disorder-based proteoforms. Cell Mol Life Sci 2019; 76:4461-4492. [PMID: 31428838 PMCID: PMC11105632 DOI: 10.1007/s00018-019-03276-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022]
Abstract
GPCR-G protein signaling system recognizes a multitude of extracellular ligands and triggers a variety of intracellular signaling cascades in response. In humans, this system includes more than 800 various GPCRs and a large set of heterotrimeric G proteins. Complexity of this system goes far beyond a multitude of pair-wise ligand-GPCR and GPCR-G protein interactions. In fact, one GPCR can recognize more than one extracellular signal and interact with more than one G protein. Furthermore, one ligand can activate more than one GPCR, and multiple GPCRs can couple to the same G protein. This defines an intricate multifunctionality of this important signaling system. Here, we show that the multifunctionality of GPCR-G protein system represents an illustrative example of the protein structure-function continuum, where structures of the involved proteins represent a complex mosaic of differently folded regions (foldons, non-foldons, unfoldons, semi-foldons, and inducible foldons). The functionality of resulting highly dynamic conformational ensembles is fine-tuned by various post-translational modifications and alternative splicing, and such ensembles can undergo dramatic changes at interaction with their specific partners. In other words, GPCRs and G proteins exist as sets of conformational/basic, inducible/modified, and functioning proteoforms characterized by a broad spectrum of structural features and possessing various functional potentials.
Collapse
Affiliation(s)
- Alexander V Fonin
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
| | - April L Darling
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Irina M Kuznetsova
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
| | - Konstantin K Turoverov
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
- Department of Biophysics, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya av. 29, St. Petersburg, 195251, Russian Federation
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow, Russian Federation.
| |
Collapse
|
11
|
Ueta Y, Yamamoto R, Kato N. Layer-specific modulation of pyramidal cell excitability by electroconvulsive shock. Neurosci Lett 2019; 709:134383. [PMID: 31325579 DOI: 10.1016/j.neulet.2019.134383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/17/2019] [Accepted: 07/16/2019] [Indexed: 10/26/2022]
Abstract
Dysregulation of cortical excitability crucially involves in behavioral and cognitive deficits of neurodegenerative and neuropsychiatric diseases. Electroconvulsive shock (ECS) changes neuronal excitability and has been used in the therapy of major depressive disorder and mood disorders. However, the action and the targets of the ECS in the cortical circuits are still poorly understood. Here we show that the ECS differently changes intrinsic properties of pyramidal cells (PCs) among superficial and deep layers. In layer 2/3 PCs, the ECS induced membrane hyperpolarization and the reduction of input resistances. In layer 5 PCs, the ECS also induced membrane hyperpolarization but had little effects on input resistances. In layer 6 PCs, the ECS had no effects on both of resting membrane potentials and input resistances. In addition, the ECS reduced the firing frequency of PCs in layer 2/3 but not in both layers 5 and 6. We further examined the ECS-induced changes in the influx of Ca2+ currents, and observed an enhanced Ca2+ currents in PCs of both layers 2/3 and 5 but not of layer 6. Thus, this study suggests the layer-specific suppression of PC excitability which will underlie the mechanism of the ECS action on the cortical activity.
Collapse
Affiliation(s)
- Yoshifumi Ueta
- Department of Physiology, Division of Neurophysiology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan.
| |
Collapse
|
12
|
Gimse K, Gorzek RC, Olin A, Osting S, Burger C. Hippocampal Homer1b/c is necessary for contextual fear conditioning and group I metabotropic glutamate receptor mediated long-term depression. Neurobiol Learn Mem 2018; 156:17-23. [PMID: 30336208 PMCID: PMC6226007 DOI: 10.1016/j.nlm.2018.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 01/24/2023]
Abstract
Coiled-coil forms of Homer1, including Homer1b and c (Homer1b/c) have been shown to play a role in hippocampal learning and memory and synaptic plasticity. We have previously found that overexpression of hippocampal Homer1c is sufficient to rescue learning and memory ability in aged learning impaired rats and in Homer1 knockout (KO) mice, and to rescue group I metabotropic glutamate receptor (mGluR1/5) mediated long-term potentiation in KO mice. Here, to determine if Homer1b/c is necessary for successful learning and memory we have utilized a rAAV5 vector expressing a Homer1b/c-targeting short hairpin RNA to knock down the expression of hippocampal Homer1b/c in adult 4-6-month old male Sprague Dawley rats. We have found that reduced hippocampal Homer1b/c expression elicits significant learning deficits in contextual fear conditioning, but not in the Morris water maze or novel object recognition tasks. Furthermore, we demonstrate that reduced hippocampal Homer1b/c is sufficient to completely block mGluR1/5 mediated long-term depression in the Schaffer collateral pathway. These results support a significant role for Homer1b/c in learning and synaptic plasticity; however, the exact role of each of these two protein isoforms in learning and memory remains elusive.
Collapse
Affiliation(s)
- Kirstan Gimse
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Medical Sciences Center, 1300 University Ave, Room 73 Bardeen, Madison, WI 53706, USA
| | - Ryan C Gorzek
- College of Letters and Science, University of Wisconsin, Madison, WI, USA
| | - Andrew Olin
- Department of Neurology, University of Wisconsin, Madison, WI, USA
| | - Sue Osting
- Department of Neurology, University of Wisconsin, Madison, WI, USA
| | - Corinna Burger
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Medical Sciences Center, 1300 University Ave, Room 73 Bardeen, Madison, WI 53706, USA; Department of Neurology, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
13
|
Zygmunt M, Piechota M, Rodriguez Parkitna J, Korostyński M. Decoding the transcriptional programs activated by psychotropic drugs in the brain. GENES BRAIN AND BEHAVIOR 2018; 18:e12511. [PMID: 30084543 DOI: 10.1111/gbb.12511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023]
Abstract
Analysis of drug-induced gene expression in the brain has long held the promise of revealing the molecular mechanisms of drug actions as well as predicting their long-term clinical efficacy. However, despite some successes, this promise has yet to be fulfilled. Here, we present an overview of the current state of understanding of drug-induced gene expression in the brain and consider the obstacles to achieving a robust prediction of the properties of psychoactive compounds based on gene expression profiles. We begin with a comprehensive overview of the mechanisms controlling drug-inducible transcription and the complexity resulting from expression of noncoding RNAs and alternative gene isoforms. Particular interest is placed on studies that examine the associations within drug classes with regard to the effects on gene transcription, alterations in cell signaling and neuropharmacological drug properties. While the ability of gene expression signatures to distinguish specific clinical classes of psychotropic and addictive drugs remains unclear, some reports show that under specific constraints, drug properties can be predicted based on gene expression. Such signatures offer a simple and effective way to classify psychotropic drugs and screen novel psychoactive compounds. Finally, we note that the amount of data regarding molecular programs activated in the brain by drug treatment has grown exponentially in recent years and that future advances may therefore come in large part from integrating the currently available high-throughput data sets.
Collapse
Affiliation(s)
- Magdalena Zygmunt
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| | - Michał Korostyński
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
14
|
Stefanik MT, Milovanovic M, Werner CT, Spainhour JCG, Wolf ME. Withdrawal From Cocaine Self-administration Alters the Regulation of Protein Translation in the Nucleus Accumbens. Biol Psychiatry 2018; 84:223-232. [PMID: 29622268 PMCID: PMC6054574 DOI: 10.1016/j.biopsych.2018.02.012] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cue-induced cocaine craving incubates during abstinence from cocaine self-administration. Expression of incubation ultimately depends on elevation of homomeric GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors in the nucleus accumbens (NAc). This adaptation requires ongoing protein translation for its maintenance. Aberrant translation is implicated in central nervous system diseases, but nothing is known about glutamatergic regulation of translation in the drug-naïve NAc or after incubation. METHODS NAc tissue was obtained from drug-naïve rats and from rats after 1 or >40 days of abstinence from extended-access cocaine or saline self-administration. Newly translated proteins were labeled using 35S-Met/Cys or puromycin. We compared basal overall translation and its regulation by metabotropic glutamate receptor 1 (mGlu1), mGlu5, and N-methyl-D-aspartate receptors (NMDARs) in drug-naïve, saline control, and cocaine rats, and we compared GluA1 and GluA2 translation by immunoprecipitating puromycin-labeled proteins. RESULTS In all groups, overall translation was unaltered by mGlu1 blockade (LY367385) but increased by mGlu5 blockade (MTEP). NMDAR blockade (AVP) increased overall translation in drug-naïve and saline control rats but not in cocaine/late withdrawal rats. Cocaine/late withdrawal rats exhibited greater translation of GluA1 (but not GluA2), which was not further affected by NMDAR blockade. CONCLUSIONS Our results suggest that increased GluA1 translation contributes to the elevated homomeric GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor levels in the NAc that mediate incubation. Additional contributions to incubation-related plasticity may result from loss of the braking influence on translation normally exerted by NMDARs. Apart from elucidating incubation-related adaptations, we found a suppressive effect of mGlu5 on NAc translation regardless of drug exposure, which is opposite to results obtained in the hippocampus and points to heterogeneity of translational regulation between brain regions.
Collapse
Affiliation(s)
- Michael T Stefanik
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - Mike Milovanovic
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - Craig T Werner
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois
| | - John C G Spainhour
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, Atlanta, Georgia
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin School of Medicine and Science, North Chicago, Illinois.
| |
Collapse
|
15
|
Lum JS, Pan B, Deng C, Huang XF, Ooi L, Newell KA. Effects of short- and long-term aripiprazole treatment on Group I mGluRs in the nucleus accumbens: Comparison with haloperidol. Psychiatry Res 2018; 260:152-157. [PMID: 29195167 DOI: 10.1016/j.psychres.2017.11.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
Abstract
The D2 receptor partial agonist, aripiprazole, has shown increased therapeutic efficacy for schizophrenia, autism and Tourette's syndrome compared to traditional antipsychotics such as the D2 receptor antagonist, haloperidol. Recent evidence suggests this superior profile may be associated with downstream effects on glutamatergic synapses. Group 1 metabotropic glutamate receptors (mGluRs) and their endogenous modulators, Norbin and Homer1, are regulated by D2 receptor activity, particularly within the nucleus accumbens (NAc), a target region of aripiprazole and haloperidol. This study sought to evaluate the effects of aripiprazole on Group 1 mGluRs, Norbin and Homer1 in the NAc, in comparison to haloperidol. Sprague-Dawley rats were orally administered daily doses of aripiprazole (2.25mg/kg), haloperidol (0.3mg/kg) or vehicle for 1 or 10-weeks. Immunoblot analyses revealed Group 1 mGluR protein levels were not altered following 1-week and 10-week aripiprazole or haloperidol treatment, compared to vehicle treated rodents. However, 1-week aripiprazole and haloperidol treatment significantly elevated Homer1a and Norbin protein expression, respectively. After 10 weeks of treatment, aripiprazole, but not haloperidol, significantly increased Norbin expression. These findings indicate the antipsychotics, aripiprazole and haloperidol, exert differential temporal effects on Norbin and Homer1 expression that may have consequences on synaptic glutamatergic transmission underlying their therapeutic profile.
Collapse
Affiliation(s)
- Jeremy S Lum
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia; Schizophrenia Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Bo Pan
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Department of Pharmacy, Medical Academy, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Chao Deng
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Xu-Feng Huang
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Lezanne Ooi
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia
| | - Kelly A Newell
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia; Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia.
| |
Collapse
|
16
|
Lee KM, Coelho MA, Sern KR, Szumlinski KK. Homer2 within the central nucleus of the amygdala modulates withdrawal-induced anxiety in a mouse model of binge-drinking. Neuropharmacology 2017; 128:448-459. [PMID: 29109058 DOI: 10.1016/j.neuropharm.2017.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/17/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023]
Abstract
A history of binge-drinking decreases protein expression of the glutamate-related scaffolding protein Homer2 within the central nucleus of the amygdala (CEA), coinciding with behavioral signs of negative affect. To assess the functional relevance of this protein change for withdrawal-induced hyper-anxiety, adult (PND 56) and adolescent (PND 28) male C57BL/6J mice were administered an intra-CEA infusion of an adeno-associated viral vector (AAV) carrying either cDNA to express Homer2 (H2-cDNA) or GFP as control. Mice underwent 14 days of binge-drinking under multi-bottle, limited-access conditions and were assayed for behavioral signs of negative affect during withdrawal using the light-dark box, marble burying, and forced swim tests (FST). Following behavioral testing, all animals experienced 5 days of drinking to evaluate the effects of prior alcohol experience and Homer2 manipulation on subsequent alcohol consumption. During protracted (4 weeks) withdrawal, adolescent alcohol-experienced GFP controls showed increased signs of negative affect across all 3 assays, compared to water-drinking GFP animals, and also showed elevated alcohol consumption during the subsequent drinking period. Homer2-cDNA infusion in adolescent-onset alcohol-drinking animals was anxiolytic and reduced subsequent alcohol consumption. Conversely, Homer2-cDNA was anxiogenic and increased drinking in water-drinking adolescents. Unfortunately, the data from adult-onset alcohol-drinking animals were confounded by low alcohol consumption and negligible behavioral signs of anxiety. Nevertheless, the present results provide novel cause-effect evidence supporting a role for CEA Homer2 in the regulation of both basal anxiety and the time-dependent intensification of negative affective states in individuals with a history of binge-drinking during adolescence.
Collapse
Affiliation(s)
- K M Lee
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, 93106-9660 CA, USA
| | - M A Coelho
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, 93106-9660 CA, USA
| | - K R Sern
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, 93106-9660 CA, USA
| | - K K Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, 93106-9660 CA, USA; Department of Molecular, Cellular and Developmental Biology, The Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, 93106-9625 CA, USA.
| |
Collapse
|
17
|
Homer1a protein expression in schizophrenia, bipolar disorder, and major depression. J Neural Transm (Vienna) 2017; 124:1261-1273. [DOI: 10.1007/s00702-017-1776-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/31/2017] [Indexed: 01/14/2023]
|
18
|
Buonaguro EF, Iasevoli F, Marmo F, Eramo A, Latte G, Avagliano C, Tomasetti C, de Bartolomeis A. Re-arrangements of gene transcripts at glutamatergic synapses after prolonged treatments with antipsychotics: A putative link with synaptic remodeling. Prog Neuropsychopharmacol Biol Psychiatry 2017; 76:29-41. [PMID: 28235555 DOI: 10.1016/j.pnpbp.2017.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 10/20/2022]
Abstract
OBJECTIVES The postsynaptic density (PSD) represents a site of dopamine-glutamate integration. Despite multiple evidence of PSD involvement in antipsychotic-induced synaptic changes, there are no direct head-to-head comparisons of the effects at the PSD of antipsychotics with different receptor profile and at different doses after chronic administration. METHODS Molecular imaging of gene expression was used to investigate whether chronic treatment with first and second generation antipsychotics (haloperidol, asenapine and olanzapine) may induce changes in the expression levels of PSD transcripts involved in schizophrenia pathophysiology, i.e. Homers, Shank1, PSD-95 and Arc. RESULTS Genes' expression patterns were differentially modulated after chronic administration of typical and atypical antipsychotics as well as by the same compound administered at different doses. Antipsychotic treatment reduced gene expression in cortical regions, while Homer1a was still induced in striatum by haloperidol even after prolonged treatment. Moreover, chronic treatments appeared to cause a "de-recruitment" of brain regions demonstrated to be activated in acute treatments, with a prominent effect in the cortex rather than in striatum. CONCLUSIONS These results let hypothesize that prolonged antipsychotic treatment may trigger a set of plastic changes involving scaffolding and effector molecules causing a possible re-arrangement of PSD transcripts in brain regions relevant to schizophrenia pathophysiology.
Collapse
Affiliation(s)
- Elisabetta Filomena Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy
| | | | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy
| | - Camilla Avagliano
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy
| | - Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Section of Psychiatry, University of Naples Federico II, Italy.
| |
Collapse
|
19
|
Abstract
The classic serotonergic hallucinogens, or psychedelics, have the ability to profoundly alter perception and behavior. These can include visual distortions, hallucinations, detachment from reality, and mystical experiences. Some psychedelics, like LSD, are able to produce these effects with remarkably low doses of drug. Others, like psilocybin, have recently been demonstrated to have significant clinical efficacy in the treatment of depression, anxiety, and addiction that persist for at least several months after only a single therapeutic session. How does this occur? Much work has recently been published from imaging studies showing that psychedelics alter brain network connectivity. They facilitate a disintegration of the default mode network, producing a hyperconnectivity between brain regions that allow centers that do not normally communicate with each other to do so. The immediate and acute effects on both behaviors and network connectivity are likely mediated by effector pathways downstream of serotonin 5-HT2A receptor activation. These acute molecular processes also influence gene expression changes, which likely influence synaptic plasticity and facilitate more long-term changes in brain neurochemistry ultimately underlying the therapeutic efficacy of a single administration to achieve long-lasting effects. In this review, we summarize what is currently known about the molecular genetic responses to psychedelics within the brain and discuss how gene expression changes may contribute to altered cellular physiology and behaviors.
Collapse
|
20
|
de Bartolomeis A, Marmo F, Buonaguro EF, Latte G, Tomasetti C, Iasevoli F. Switching antipsychotics: Imaging the differential effect on the topography of postsynaptic density transcripts in antipsychotic-naïve vs. antipsychotic-exposed rats. Prog Neuropsychopharmacol Biol Psychiatry 2016; 70:24-38. [PMID: 27177972 DOI: 10.1016/j.pnpbp.2016.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/17/2016] [Accepted: 04/27/2016] [Indexed: 10/24/2022]
Abstract
The postsynaptic density (PSD) has been regarded as a functional switchboard at the crossroads of a dopamine-glutamate interaction, and it is putatively involved in the pathophysiology of psychosis. Indeed, it has been demonstrated that antipsychotics may modulate several PSD transcripts, such as PSD-95, Shank, and Homer. Despite switching antipsychotics is a frequent strategy to counteract lack of efficacy and/or side effect onset in clinical practice, no information is available on the effects of sequential treatments with different antipsychotics on PSD molecules. The aim of this study was to evaluate whether a previous exposure to a typical antipsychotic and a switch to an atypical one may affect the expression of PSD transcripts, in order to evaluate potential neurobiological correlates of this common clinical practice, with specific regards to putative synaptic plasticity processes. We treated male Sprague-Dawley rats intraperitoneally for 15days with haloperidol or vehicle, then from the sixteenth day we switched the animals to amisulpride or continued to treat them with vehicle or haloperidol for 15 additional days. In this way we got six first treatment/second treatment groups: vehicle/vehicle, vehicle/haloperidol, vehicle/amisulpride, haloperidol/vehicle, haloperidol/haloperidol, haloperidol/amisulpride. In this paradigm, we evaluated the expression of brain transcripts belonging to relevant and interacting PSD proteins, both of the Immediate-Early Gene (Homer1a, Arc) and the constitutive classes (Homer1b/c and PSD-95). The major finding was the differential effect of amisulpride on gene transcripts when administered in naïve vs. antipsychotic-pretreated rats, with modifications of the ratio between Homer1a/Homer1b transcripts and differential effects in cortex and striatum. These results suggest that the neurobiological effects on PSD transcripts of amisulpride, and possibly of other antipsychotics, may be greatly affected by prior antipsychotic treatments and may impact significantly on the switching procedure.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy.
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| |
Collapse
|
21
|
Molecular evidence of synaptic pathology in the CA1 region in schizophrenia. NPJ SCHIZOPHRENIA 2016; 2:16022. [PMID: 27430010 PMCID: PMC4944906 DOI: 10.1038/npjschz.2016.22] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 11/09/2022]
Abstract
Alterations of postsynaptic density (PSD)95-complex proteins in schizophrenia ostensibly induce deficits in synaptic plasticity, the molecular process underlying cognitive functions. Although some PSD95-complex proteins have been previously examined in the hippocampus in schizophrenia, the status of other equally important molecules is unclear. This is especially true in the cornu ammonis (CA)1 hippocampal subfield, a region that is critically involved in the pathophysiology of the illness. We thus performed a quantitative immunoblot experiment to examine PSD95 and several of its associated proteins in the CA1 region, using post mortem brain samples derived from schizophrenia subjects with age-, sex-, and post mortem interval-matched controls (n=20/group). Our results indicate a substantial reduction in PSD95 protein expression (-61.8%). Further analysis showed additional alterations to the scaffold protein Homer1 (Homer1a: +42.9%, Homer1b/c: -24.6%), with a twofold reduction in the ratio of Homer1b/c:Homer1a isoforms (P=0.011). Metabotropic glutamate receptor 1 (mGluR1) protein levels were significantly reduced (-32.7%), and Preso, a protein that supports interactions between Homer1 or PSD95 with mGluR1, was elevated (+83.3%). Significant reduction in synaptophysin (-27.8%) was also detected, which is a validated marker of synaptic density. These findings support the presence of extensive molecular abnormalities to PSD95 and several of its associated proteins in the CA1 region in schizophrenia, offering a small but significant step toward understanding how proteins in the PSD are altered in the schizophrenia brain, and their relevance to overall hippocampal and cognitive dysfunction in the illness.
Collapse
|
22
|
Abstract
UNLABELLED The possibility that mechanisms of synaptic modulation differ between males and females has far-reaching implications for understanding brain disorders that vary between the sexes. We found recently that 17β-estradiol (E2) acutely suppresses GABAergic inhibition in the hippocampus of female rats through a sex-specific estrogen receptor α (ERα), mGluR, and endocannabinoid-dependent mechanism. Here, we define the intracellular signaling that links ERα, mGluRs, and endocannabinoids in females and identify where in this pathway males and females differ. Using a combination of whole-cell patch-clamp recording and biochemical analyses in hippocampal slices from young adult rats, we show that E2 acutely suppresses inhibition in females through mGluR1 stimulation of phospholipase C, leading to inositol triphosphate (IP3) generation, activation of the IP3 receptor (IP3R), and postsynaptic endocannabinoid release, likely of anandamide. Analysis of sex differences in this pathway showed that E2 stimulates a much greater increase in IP3 levels in females than males, whereas the group I mGluR agonist DHPG increases IP3 levels equivalently in each sex. Coimmunoprecipitation showed that ERα-mGluR1 and mGluR1-IP3R complexes exist in both sexes but are regulated by E2 only in females. Independently of E2, a fatty acid amide hydrolase inhibitor, which blocks breakdown of anandamide, suppressed >50% of inhibitory synapses in females with no effect in males, indicating tonic endocannabinoid release in females that is absent in males. Together, these studies demonstrate sex differences in both E2-dependent and E2-independent regulation of the endocannabinoid system and suggest that manipulation of endocannabinoids in vivo could affect physiological and behavioral responses differently in each sex. SIGNIFICANCE STATEMENT Many brain disorders vary between the sexes, yet the degree to which this variation arises from differential experience versus intrinsic biological sex differences is unclear. In this study, we demonstrate intrinsic sex differences in molecular regulation of a key neuromodulatory system, the endocannabinoid system, in the hippocampus. Endocannabinoids are involved in diverse aspects of physiology and behavior that involve the hippocampus, including cognitive and motivational state, responses to stress, and neurological disorders such as epilepsy. Our finding that molecular regulation of the endocannabinoid system differs between the sexes suggests mechanisms through which experiences or therapeutics that engage endocannabinoids could affect males and females differently.
Collapse
|
23
|
de Bartolomeis A, Errico F, Aceto G, Tomasetti C, Usiello A, Iasevoli F. D-aspartate dysregulation in Ddo(-/-) mice modulates phencyclidine-induced gene expression changes of postsynaptic density molecules in cortex and striatum. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:35-43. [PMID: 25979765 DOI: 10.1016/j.pnpbp.2015.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
N-methyl-D-aspartate receptor (NMDAR) hypofunction has been considered a key alteration in schizophrenia pathophysiology. Thus, several strategies aimed at enhancing glutamatergic transmission, included the introduction in therapy of D-amino acids, such as D-serine and D-cycloserine augmentation, have been proposed to counteract difficult-to-treat symptoms or treatment-resistant forms of schizophrenia. Another D-amino acid, D-aspartate, has recently gained increasing interest for its role in NMDAR activation and has been found reduced in post-mortem cortex of schizophrenia patients. NMDAR is the core of the postsynaptic density (PSD), a postsynaptic site involved in glutamate signaling and responsive to antipsychotic treatment. In this study, we investigated striatal and cortical gene expression of key PSD transcripts (i.e. Homer1a, Homer1b/c, and PSD-95) in mice with persistently elevated brain D-aspartate-levels, i.e. the D-aspartate-oxidase knockout mice (Ddo(-/-)). These animal models were analyzed both in naive condition and after phencyclidine (PCP) treatment. Naive Ddo(-/-) mice showed decreased Homer1a expression in the prefrontal cortex, increased Homer1b/c expression in the striatum, and decreased PSD-95 expression in the striatum and in the cortex. Acute PCP treatment restored, and even potentiated, Homer1a expression in the prefrontal cortex of mutant mice, while it had limited effects on the other genes. These results suggest that persistently elevated D-aspartate, by enhancing NMDA transmission, may cause complex adaptive mechanisms affecting Homer1a, which in turn may explain the recently demonstrated protective effects of this D-amino acid against PCP-induced behavioral alterations, such as ataxic behavior.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy.
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy
| |
Collapse
|
24
|
Wang Y, Rao W, Zhang C, Zhang C, Liu MD, Han F, Yao LB, Han H, Luo P, Su N, Fei Z. Scaffolding protein Homer1a protects against NMDA-induced neuronal injury. Cell Death Dis 2015; 6:e1843. [PMID: 26247728 PMCID: PMC4558508 DOI: 10.1038/cddis.2015.216] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/19/2015] [Accepted: 06/25/2015] [Indexed: 01/18/2023]
Abstract
Excessive N-methyl-D-aspartate receptor (NMDAR) activation and the resulting activation of neuronal nitric oxide synthase (nNOS) cause neuronal injury. Homer1b/c facilitates NMDAR-PSD95-nNOS complex interactions, and Homer1a is a negative competitor of Homer1b/c. We report that Homer1a was both upregulated by and protected against NMDA-induced neuronal injury in vitro and in vivo. The neuroprotective activity of Homer1a was associated with NMDA-induced Ca2+ influx, oxidative stress and the resultant downstream signaling activation. Additionally, we found that Homer1a functionally regulated NMDAR channel properties in neurons, but did not regulate recombinant NR1/NR2B receptors in HEK293 cells. Furthermore, we found that Homer1a detached the physical links among NR2B, PSD95 and nNOS and reduced the membrane distribution of NMDAR. NMDA-induced neuronal injury was more severe in Homer1a homozygous knockout mice (KO, Homer1a−/−) when compared with NMDA-induced neuronal injury in wild-type mice (WT, Homer1a+/+). Additionally, Homer1a overexpression in the cortex of Homer1a−/− mice alleviated NMDA-induced neuronal injury. These findings suggest that Homer1a may be a key neuroprotective endogenous molecule that protects against NMDA-induced neuronal injury by disassembling NR2B-PSD95-nNOS complexes and reducing the membrane distribution of NMDARs.
Collapse
Affiliation(s)
- Y Wang
- 1] Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China [2] Department of Neurosurgery, Wuhan Zhong Xin Hospital, Wuhan, P.R. China
| | - W Rao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| | - C Zhang
- Department of Neurology, Second Artillery General Hospital of PLA, Beijing, P.R. China
| | - C Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| | - M-D Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| | - F Han
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| | - L-b Yao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, P.R. China
| | - H Han
- Department of Medical Genetics and Developmental Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, P.R. China
| | - P Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| | - N Su
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| | - Z Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, P.R. China
| |
Collapse
|
25
|
Protein expression of targets of the FMRP regulon is altered in brains of subjects with schizophrenia and mood disorders. Schizophr Res 2015; 165:201-11. [PMID: 25956630 PMCID: PMC5037955 DOI: 10.1016/j.schres.2015.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 03/25/2015] [Accepted: 04/09/2015] [Indexed: 12/31/2022]
Abstract
Fragile X mental retardation protein (FMRP) is an RNA binding protein with 842 target mRNAs in mammalian brain. Silencing of the fragile X mental retardation 1 (FMR1) gene leads to loss of expression of FMRP and upregulated metabotropic glutamate receptor 5 (mGluR5) signaling resulting in the multiple physical and cognitive deficits associated with fragile X syndrome (FXS). Reduced FMRP expression has been identified in subjects with autism, schizophrenia, bipolar disorder, and major depression who do not carry the mutation for FMR1. Our laboratory has recently demonstrated altered expression of four downstream targets of FMRP-mGluR5 signaling in brains of subjects with autism: homer 1, amyloid beta A4 precursor protein (APP), ras-related C3 botulinum toxin substrate 1 (RAC1), and striatal-enriched protein tyrosine phosphatase (STEP). In the current study we investigated the expression of the same four proteins in lateral cerebella of subjects with schizophrenia, bipolar disorder, and major depression and in frontal cortex of subjects with schizophrenia and bipolar disorder. In frontal cortex we observed: 1) reduced expression of 120 kDa form of APP in subjects with schizophrenia and bipolar disorder; 2) reduced expression of 61 kDa and 33k Da forms of STEP in subjects with schizophrenia; 3) reduced expression of 88 kDa form of APP in subjects with bipolar disorder; and 3) trends for reduced expression of 88 kDa form of APP and homer 1 in subjects with schizophrenia and bipolar disorder, respectively. In lateral cerebella there was no group difference, however we observed increased expression of RAC1 in subjects with bipolar disorder, and trends for increased RAC1 in subjects with schizophrenia and major depression. Our results provide further evidence that proteins involved in the FMRP-mGluR5 signaling pathway are altered in schizophrenia and mood disorders.
Collapse
|
26
|
Wagner KV, Hartmann J, Labermaier C, Häusl AS, Zhao G, Harbich D, Schmid B, Wang XD, Santarelli S, Kohl C, Gassen NC, Matosin N, Schieven M, Webhofer C, Turck CW, Lindemann L, Jaschke G, Wettstein JG, Rein T, Müller MB, Schmidt MV. Homer1/mGluR5 activity moderates vulnerability to chronic social stress. Neuropsychopharmacology 2015; 40:1222-33. [PMID: 25409593 PMCID: PMC4367467 DOI: 10.1038/npp.2014.308] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 12/28/2022]
Abstract
Stress-induced psychiatric disorders, such as depression, have recently been linked to changes in glutamate transmission in the central nervous system. Glutamate signaling is mediated by a range of receptors, including metabotropic glutamate receptors (mGluRs). In particular, mGluR subtype 5 (mGluR5) is highly implicated in stress-induced psychopathology. The major scaffold protein Homer1 critically interacts with mGluR5 and has also been linked to several psychopathologies. Yet, the specific role of Homer1 in this context remains poorly understood. We used chronic social defeat stress as an established animal model of depression and investigated changes in transcription of Homer1a and Homer1b/c isoforms and functional coupling of Homer1 to mGluR5. Next, we investigated the consequences of Homer1 deletion, overexpression of Homer1a, and chronic administration of the mGluR5 inverse agonist CTEP (2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine) on the effects of chronic stress. In mice exposed to chronic stress, Homer1b/c, but not Homer1a, mRNA was upregulated and, accordingly, Homer1/mGluR5 coupling was disrupted. We found a marked hyperactivity behavior as well as a dysregulated hypothalamic-pituitary-adrenal axis activity in chronically stressed Homer1 knockout (KO) mice. Chronic administration of the selective and orally bioavailable mGluR5 inverse agonist, CTEP, was able to recover behavioral alterations induced by chronic stress, whereas overexpression of Homer1a in the hippocampus led to an increased vulnerability to chronic stress, reflected in an increased physiological response to stress as well as enhanced depression-like behavior. Overall, our results implicate the glutamatergic system in the emergence of stress-induced psychiatric disorders, and support the Homer1/mGluR5 complex as a target for the development of novel antidepressant agents.
Collapse
Affiliation(s)
- Klaus V Wagner
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jakob Hartmann
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Christiana Labermaier
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alexander S Häusl
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Gengjing Zhao
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Daniela Harbich
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bianca Schmid
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Sara Santarelli
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Christine Kohl
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Nils C Gassen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Natalie Matosin
- Faculty of Science, Medicine and Health and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia,Schizophrenia Research Institute, Sydney NSW, Australia
| | - Marcel Schieven
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Christian Webhofer
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Christoph W Turck
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lothar Lindemann
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology, and Rare Diseases Translational Area (NORD), Basel, Switzerland
| | - Georg Jaschke
- Roche Pharmaceutical Research and Early Development, Discovery Chemistry, Basel, Switzerland
| | - Joseph G Wettstein
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology, and Rare Diseases Translational Area (NORD), Basel, Switzerland
| | - Theo Rein
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Marianne B Müller
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mathias V Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany, Tel: +49 89 30622 519, Fax: +49 89 30622 610, E-mail:
| |
Collapse
|
27
|
Azaiez H, Decker AR, Booth KT, Simpson AC, Shearer AE, Huygen PLM, Bu F, Hildebrand MS, Ranum PT, Shibata SB, Turner A, Zhang Y, Kimberling WJ, Cornell RA, Smith RJH. HOMER2, a stereociliary scaffolding protein, is essential for normal hearing in humans and mice. PLoS Genet 2015; 11:e1005137. [PMID: 25816005 PMCID: PMC4376867 DOI: 10.1371/journal.pgen.1005137] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/10/2015] [Indexed: 12/29/2022] Open
Abstract
Hereditary hearing loss is a clinically and genetically heterogeneous disorder. More than 80 genes have been implicated to date, and with the advent of targeted genomic enrichment and massively parallel sequencing (TGE+MPS) the rate of novel deafness-gene identification has accelerated. Here we report a family segregating post-lingual progressive autosomal dominant non-syndromic hearing loss (ADNSHL). After first excluding plausible variants in known deafness-causing genes using TGE+MPS, we completed whole exome sequencing in three hearing-impaired family members. Only a single variant, p.Arg185Pro in HOMER2, segregated with the hearing-loss phenotype in the extended family. This amino acid change alters a highly conserved residue in the coiled-coil domain of HOMER2 that is essential for protein multimerization and the HOMER2-CDC42 interaction. As a scaffolding protein, HOMER2 is involved in intracellular calcium homeostasis and cytoskeletal organization. Consistent with this function, we found robust expression in stereocilia of hair cells in the murine inner ear and observed that over-expression of mutant p.Pro185 HOMER2 mRNA causes anatomical changes of the inner ear and neuromasts in zebrafish embryos. Furthermore, mouse mutants homozygous for the targeted deletion of Homer2 present with early-onset rapidly progressive hearing loss. These data provide compelling evidence that HOMER2 is required for normal hearing and that its sequence alteration in humans leads to ADNSHL through a dominant-negative mode of action. The most frequent sensory disorder worldwide is hearing impairment. It impacts over 5% of the world population (360 million persons), and is characterized by extreme genetic heterogeneity. Over 80 genes have been implicated in isolated (also referred to as ‘non-syndromic’) hearing loss, and abundant evidence supports the existence of many more ‘deafness-causing’ genes. In this study, we used a sequential screening strategy to first exclude causal mutations in known deafness-causing genes in a family segregating autosomal dominant non-syndromic hearing loss. We next turned to whole exome sequencing and identified a single variant—p.Arg185Pro in HOMER2—that segregated with the phenotype in the extended family. To validate the pathological significance of this mutation, we studied two animal models. In zebrafish, we overexpressed mutant HOMER2 and observed inner ear defects; and in mice we documented robust expression in stereocilia of cochlear hair cells and demonstrated that its absence causes early-onset progressive deafness. Our data offer novel insights into gene pathways essential for normal auditory function and the maintenance of cochlear hair cells.
Collapse
Affiliation(s)
- Hela Azaiez
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Amanda R. Decker
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Kevin T. Booth
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Allen C. Simpson
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - A. Eliot Shearer
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Patrick L. M. Huygen
- Department of Otorhinolaryngology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Fengxiao Bu
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Michael S. Hildebrand
- Austin Health, Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Paul T. Ranum
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Seiji B. Shibata
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Ann Turner
- Self-employed physician, Menlo Park, California, United States of America
| | - Yuzhou Zhang
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - William J. Kimberling
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
| | - Robert A. Cornell
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Richard J. H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology University of Iowa, Iowa City, Iowa, United States of America
- Interdepartmental PhD Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
28
|
Homer protein-metabotropic glutamate receptor binding regulates endocannabinoid signaling and affects hyperexcitability in a mouse model of fragile X syndrome. J Neurosci 2015; 35:3938-45. [PMID: 25740522 DOI: 10.1523/jneurosci.4499-14.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The Fmr1 knock-out mouse model of fragile X syndrome (Fmr1(-/y)) has an epileptogenic phenotype that is triggered by group I metabotropic glutamate receptor (mGluR) activation. We found that a membrane-permeable peptide that disrupts mGluR5 interactions with long-form Homers enhanced mGluR-induced epileptiform burst firing in wild-type (WT) animals, replicating the early stages of hyperexcitability in Fmr1(-/y). The peptide enhanced mGluR-evoked endocannabinoid (eCB)-mediated suppression of inhibitory synapses, decreased it at excitatory synapses in WTs, but had no effect on eCB actions in Fmr1(-/y). At a low concentration, the mGluR agonist did not generate eCBs at excitatory synapses but nevertheless induced burst firing in both Fmr1(-/y) and peptide-treated WT slices. This burst firing was suppressed by a cannabinoid receptor antagonist. We suggest that integrity of Homer scaffolds is essential for normal mGluR-eCB functioning and that aberrant eCB signaling resulting from disturbances of this molecular structure contributes to the epileptic phenotype of Fmr1(-/y).
Collapse
|
29
|
Cavarsan CF, Matsuo A, Blanco MM, Mello LE. Maximal electroshock-induced seizures are able to induce Homer1a mRNA expression but not pentylenetetrazole-induced seizures. Epilepsy Behav 2015; 44:90-5. [PMID: 25659045 DOI: 10.1016/j.yebeh.2014.12.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Homer1a is a protein that regulates metabotropic glutamate receptors involved in neural plasticity processes. Recently, we demonstrated that Homer1a mRNA is enhanced after pilocarpine-induced status epilepticus. Here, we investigated whether a single acute seizure triggered by means of pentylenetetrazole (PTZ) injection or maximal electroshock (MES) stimulation (2 different seizure models) would alter Homer1a expression in the hippocampus. METHODS Male Wistar rats subjected to the PTZ or MES model were analyzed 2h, 8h, 24h, and 7days after seizure induction. Homer1a, mGluR1, and mGluR5 mRNA expression levels in hippocampal extracts were analyzed by quantitative PCR. RESULTS Quantitative PCR revealed Homer1a overexpression at 2h after MES-induced tonic-clonic seizures compared to control, but the overexpression did not remain elevated after 8h. Pentylenetetrazole-induced seizures, in contrast, were not able to change Homer1a mRNA expression. No differences were observed at these time points after seizures for mGluR1 and mGluR5 mRNA expression in any of the models. SIGNIFICANCE Our data indicate that the levels of Homer1a mRNA were transiently increased only after MES-induced tonic-clonic seizures (and not after PTZ-induced seizures). We suggest that Homer1a expression may be dependent on seizure intensity or on specific brain circuit activation. We suggest that Homer1a may contribute to counteract hyperexcitability processes.
Collapse
Affiliation(s)
- Clarissa F Cavarsan
- Department of Physiology, Universidade Federal de São Paulo, Pedro de Toledo St, 669, 3rd floor, 04039-032 São Paulo, SP, Brazil
| | - Alisson Matsuo
- UNONEX, Department of Microbiology, Immunology, and Parasitology, Universidade Federal de São Paulo, Building Prof. Dr. Antonio C. Mattos Paiva, Botucatu St, 862, 8th floor, Vila Clementino, 04023-062 São Paulo, SP, Brazil
| | - Miriam M Blanco
- Department of Physiology, Universidade Federal de São Paulo, Pedro de Toledo St, 669, 3rd floor, 04039-032 São Paulo, SP, Brazil
| | - Luiz E Mello
- Department of Physiology, Universidade Federal de São Paulo, Pedro de Toledo St, 669, 3rd floor, 04039-032 São Paulo, SP, Brazil.
| |
Collapse
|
30
|
Huang W, Liu X, Fei Z, Zhang Y, Yang J. Down-regulation of Homer1b/c expression protects cultured neurons after traumatic injury. Neural Regen Res 2014; 7:2176-81. [PMID: 25538737 PMCID: PMC4268715 DOI: 10.3969/j.issn.1673-5374.2012.028.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 09/24/2012] [Indexed: 11/18/2022] Open
Abstract
Activation of metabotropic glutamate receptor 1a aggravates traumatic brain injury. The constitutively expressed protein Homer1b/c participates in delivering and anchoring metabotropic glutamate receptors in neurons. Here, we aimed to verify whether down-regulation of Homer1b/c by RNA interference could protect cultured rat cortical neurons from traumatic injury. We showed that 36 hours after transfection of Homer1b/c small interfering RNA, metabotropic glutamate receptor 1a was present only in the neuronal cytoplasm, but not in the dendrites. Calcium fluorescence intensity was also decreased significantly. Moreover, lactate dehydrogenase concentration was significantly decreased in Homer1b/c small interfering RNA-transfected cells compared with that in untransfected and control small interfering RNA-transfected cells 24 hours after traumatic neuronal injury. Our findings indicate that down-regulation of Homer1b/c could reduce metabotropic glutamate receptor 1a transfer from the cell body to the dendrite, relieve calcium overload, and protect neurons from traumatic injury.
Collapse
Affiliation(s)
- Weidong Huang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Xiaobin Liu
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University of Chinese PLA, Xi'an 710032, Shaanxi Province, China
| | - Yuelin Zhang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| | - Jun Yang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
| |
Collapse
|
31
|
Jong YJI, Sergin I, Purgert CA, O'Malley KL. Location-dependent signaling of the group 1 metabotropic glutamate receptor mGlu5. Mol Pharmacol 2014; 86:774-85. [PMID: 25326002 PMCID: PMC4244594 DOI: 10.1124/mol.114.094763] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
Although G protein-coupled receptors are primarily known for converting extracellular signals into intracellular responses, some receptors, such as the group 1 metabotropic glutamate receptor, mGlu5, are also localized on intracellular membranes where they can mediate both overlapping and unique signaling effects. Thus, besides "ligand bias," whereby a receptor's signaling modality can shift from G protein dependence to independence, canonical mGlu5 receptor signaling can also be influenced by "location bias" (i.e., the particular membrane and/or cell type from which it signals). Because mGlu5 receptors play important roles in both normal development and in disorders such as Fragile X syndrome, autism, epilepsy, addiction, anxiety, schizophrenia, pain, dyskinesias, and melanoma, a large number of drugs are being developed to allosterically target this receptor. Therefore, it is critical to understand how such drugs might be affecting mGlu5 receptor function on different membranes and in different brain regions. Further elucidation of the site(s) of action of these drugs may determine which signal pathways mediate therapeutic efficacy.
Collapse
Affiliation(s)
- Yuh-Jiin I Jong
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Ismail Sergin
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Carolyn A Purgert
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Karen L O'Malley
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
32
|
Iasevoli F, Buonaguro EF, Sarappa C, Marmo F, Latte G, Rossi R, Eramo A, Tomasetti C, de Bartolomeis A. Regulation of postsynaptic plasticity genes' expression and topography by sustained dopamine perturbation and modulation by acute memantine: relevance to schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54:299-314. [PMID: 25025505 DOI: 10.1016/j.pnpbp.2014.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/26/2014] [Accepted: 07/03/2014] [Indexed: 11/25/2022]
Abstract
A relevant role for dopamine-glutamate interaction has been reported in the pathophysiology and treatment of psychoses. Dopamine and glutamate may interact at multiple levels, including the glutamatergic postsynaptic density (PSD), an electron-dense thickening that has gained recent attention as a switchboard of dopamine-glutamate interactions and for its role in synaptic plasticity. Recently, glutamate-based strategies, such as memantine add-on to antipsychotics, have been proposed for refractory symptoms of schizophrenia, e.g. cognitive impairment. Both antipsychotics and memantine regulate PSD transcripts but sparse information is available on memantine's effects under dopamine perturbation. We tested gene expression changes of the Homer1 and PSD-95 PSD proteins in models of sustained dopamine perturbation, i.e. subchronic treatment by: a) GBR-12909, a dopamine receptor indirect agonist; b) haloperidol, a D2R antagonist; c) SCH-23390, a dopamine D1 receptor (D1R) antagonist; and d) SCH-23390+haloperidol. On the last day of treatment, rats were acutely treated with vehicle or memantine. The Homer1a immediate-early gene was significantly induced by haloperidol and by haloperidol+SCH-23390. The gene was not induced by SCH-23390 per se or by GBR-12909. Expression of the constitutive genes Homer1b/c and PSD-95 was less affected by these dopaminergic paradigms. Acute memantine administration significantly increased Homer1a expression by the dopaminergic compounds used herein. Both haloperidol and haloperidol+SCH-23390 shifted Homer1a/Homer1b/c ratio of expression toward Homer1a. This pattern was sharpened by acute memantine. Dopaminergic compounds and acute memantine also differentially affected topographic distribution of gene expression and coordinated expression of Homer1a among cortical-subcortical regions. These results indicate that dopaminergic perturbations may affect glutamatergic signaling in different directions. Memantine may help partially revert dopamine-mediated glutamatergic dysfunctions.
Collapse
Affiliation(s)
- Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Chiara Sarappa
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Rodolfo Rossi
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Anna Eramo
- Medical Affairs & Phase IV Clinical Affairs, Lundbeck Pharmaceutical Services LLC, Deerfield, IL, United States
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy
| | - Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy.
| |
Collapse
|
33
|
Pomierny-Chamioło L, Rup K, Pomierny B, Niedzielska E, Kalivas PW, Filip M. Metabotropic glutamatergic receptors and their ligands in drug addiction. Pharmacol Ther 2014; 142:281-305. [DOI: 10.1016/j.pharmthera.2013.12.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 02/07/2023]
|
34
|
Iasevoli F, Tomasetti C, Buonaguro EF, de Bartolomeis A. The glutamatergic aspects of schizophrenia molecular pathophysiology: role of the postsynaptic density, and implications for treatment. Curr Neuropharmacol 2014; 12:219-38. [PMID: 24851087 PMCID: PMC4023453 DOI: 10.2174/1570159x12666140324183406] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/14/2014] [Accepted: 03/14/2014] [Indexed: 01/23/2023] Open
Abstract
Schizophrenia is one of the most debilitating psychiatric diseases with a lifetime prevalence of approximately
1%. Although the specific molecular underpinnings of schizophrenia are still unknown, evidence has long linked its
pathophysiology to postsynaptic abnormalities.
The postsynaptic density (PSD) is among the molecular structures suggested to be potentially involved in schizophrenia.
More specifically, the PSD is an electron-dense thickening of glutamatergic synapses, including ionotropic and
metabotropic glutamate receptors, cytoskeletal and scaffolding proteins, and adhesion and signaling molecules. Being
implicated in the postsynaptic signaling of multiple neurotransmitter systems, mostly dopamine and glutamate, the PSD
constitutes an ideal candidate for studying dopamine-glutamate disturbances in schizophrenia. Recent evidence suggests
that some PSD proteins, such as PSD-95, Shank, and Homer are implicated in severe behavioral disorders, including
schizophrenia. These findings, further corroborated by genetic and animal studies of schizophrenia, offer new insights for
the development of pharmacological strategies able to overcome the limitations in terms of efficacy and side effects of
current schizophrenia treatment. Indeed, PSD proteins are now being considered as potential molecular targets against this
devastating illness.
The current paper reviews the most recent hypotheses on the molecular mechanisms underlying schizophrenia
pathophysiology. First, we review glutamatergic dysfunctions in schizophrenia and we provide an update on postsynaptic
molecules involvement in schizophrenia pathophysiology by addressing both human and animal studies. Finally, the
possibility that PSD proteins may represent potential targets for new molecular interventions in psychosis will be
discussed.
Collapse
Affiliation(s)
- Felice Iasevoli
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| | - Andrea de Bartolomeis
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| |
Collapse
|
35
|
Protective effect of Homer 1a on tumor necrosis factor-α with cycloheximide-induced apoptosis is mediated by mitogen-activated protein kinase pathways. Apoptosis 2013; 17:975-88. [PMID: 22660975 DOI: 10.1007/s10495-012-0736-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Although Homer 1, of the postsynaptic density, regulates apoptosis, the signaling mechanisms are not fully elucidated. In this study, we found that tumor necrosis factor-α (TNF-α)/cycloheximide (CHX) treatment transiently increased Homer 1a (the short variant of Homer 1), but did not affect Homer 1b/c (the long variant of Homer 1). Overexpression of Homer 1a blocked TNF-α/CHX-induced apoptotic cell death, whereas inhibition of Homer 1a induction enhanced the pro-apoptotic effect of TNF-α/CHX treatment. Moreover, brain-derived neurotrophic factor, as a potential activator of endogenous Homer 1a, inhibited apoptotic cell death after TNF-α/CHX treatment through induction of Homer 1a. Since three major mitogen-activated protein kinase (MAPK) pathways have important roles in apoptosis, we examined if Homer 1a is involved in the effects of MAPK pathways on apoptosis. It was shown that inhibition of the ERK1/2 pathway increased the expression and the protective effect of Homer 1a, but inhibition of the p38 pathway produced the opposite effect. Cross-talk among MAPK pathways was also associated with the regulation of Homer 1a during apoptotic cell death. Blocking the p38 pathway increased the activity in the ERK1/2 pathway, while inhibition of ERK1/2 pathway abolished the effect of p38 inhibitor on Homer 1a. Furthermore, Homer 1a reversely affected the activation of MAPK pathways. These findings suggest that Homer 1a plays an important role in the prevention of apoptotic cell death and contributes to distinct regulatory effects of MAPK pathways on apoptotic cell death.
Collapse
|
36
|
Synaptic depression via mGluR1 positive allosteric modulation suppresses cue-induced cocaine craving. Nat Neurosci 2013; 17:73-80. [PMID: 24270186 PMCID: PMC3971923 DOI: 10.1038/nn.3590] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/31/2013] [Indexed: 01/25/2023]
Abstract
Cue-induced cocaine craving is a major cause of relapse in abstinent addicts. In rats, cue-induced craving progressively intensifies (incubates) during withdrawal from extended-access cocaine self-administration. After ~1 month of withdrawal, incubated craving is mediated by Ca2+-permeable AMPARs (CP-AMPARs) that accumulate in the nucleus accumbens (NAc). We found that decreased mGluR1 surface expression in the NAc precedes and enables CP-AMPAR accumulation. Thus, restoring mGluR1 tone by administering repeated injections of an mGluR1 positive allosteric modulator (PAM) prevented CP-AMPAR accumulation and incubation, whereas blocking mGluR1 transmission at even earlier withdrawal times accelerated CP-AMPAR accumulation. In studies conducted after prolonged withdrawal, when CP-AMPAR levels and cue-induced craving are high, we found that systemic administration of an mGluR1 PAM attenuated the expression of incubated craving by reducing CP-AMPAR transmission in the NAc to control levels. These results demonstrate a strategy whereby recovering addicts could use a systemically active compound to protect against cue-induced relapse.
Collapse
|
37
|
de Bartolomeis A, Sarappa C, Buonaguro EF, Marmo F, Eramo A, Tomasetti C, Iasevoli F. Different effects of the NMDA receptor antagonists ketamine, MK-801, and memantine on postsynaptic density transcripts and their topography: role of Homer signaling, and implications for novel antipsychotic and pro-cognitive targets in psychosis. Prog Neuropsychopharmacol Biol Psychiatry 2013; 46:1-12. [PMID: 23800465 DOI: 10.1016/j.pnpbp.2013.06.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/10/2013] [Accepted: 06/14/2013] [Indexed: 12/15/2022]
Abstract
Administration of NMDA receptor antagonists, such as ketamine and MK-801, may induce psychotic-like behaviors in preclinical models of schizophrenia. Ketamine has also been observed to exacerbate psychotic symptoms in schizophrenia patients. However, memantine, a non-competitive NMDA receptor antagonist approved for Alzheimer's disease and proposed for antipsychotic augmentation, may challenge this view. To date, the molecular mechanisms by which these NMDA receptor antagonists cause different neurochemical, behavioral, and clinical effects are still a matter of debate. Here, we investigated by molecular imaging whether these agents could differently modulate gene expression and topographical distribution of glutamatergic postsynaptic density (PSD) proteins. We focused on Homer1a/Homer1b/PSD-95 signaling network, which may be implicated in glutamate-dependent synaptic plasticity, as well as in psychosis pathophysiology and treatment. Ketamine (25 and 50mg/kg) and MK-801 (0.8mg/kg) significantly induced the transcripts of immediate-early genes (Arc, c-fos, and Homer1a) in cortical regions compared to vehicle, whereas they reduced Homer1b and PSD-95 expression in cortical and striatal regions. Differently, memantine (5mg/kg) did not increase Homer1a signal compared to vehicle, whereas it induced c-fos in the somatosensory and in the medial agranular cortices. Moreover, memantine did not affect Homer1b and PSD-95 expression. When compared to ketamine and MK-801, memantine significantly increased the expression of c-fos, Homer1b and PSD-95. Overall, ketamine and MK-801 prominently increased Homer1a/Homer1b expression ratio, whereas memantine elicited the opposite effect. These data may support the view that ketamine, MK-801 and memantine exert divergent effects on PSD transcripts, which may contribute to their partially different behavioral and clinical effects.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
38
|
Imbalances in prefrontal cortex CC-Homer1 versus CC-Homer2 expression promote cocaine preference. J Neurosci 2013; 33:8101-13. [PMID: 23658151 DOI: 10.1523/jneurosci.1727-12.2013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Homer postsynaptic scaffolding proteins regulate forebrain glutamate transmission and thus, are likely molecular candidates mediating hypofrontality in addiction. Protracted withdrawal from cocaine experience increases the relative expression of Homer2 versus Homer1 isoforms within medial prefrontal cortex (mPFC). Thus, this study used virus-mediated gene transfer strategies to investigate the functional relevance of an imbalance in mPFC Homer1/2 expression as it relates to various measures of sensorimotor, cognitive, emotional and motivational processing, as well as accompanying alterations in extracellular glutamate in C57BL/6J mice. mPFC Homer2b overexpression elevated basal glutamate content and blunted cocaine-induced glutamate release within the mPFC, whereas Homer2b knockdown produced the opposite effects. Despite altering mPFC glutamate, Homer2b knockdown failed to influence cocaine-elicited conditioned place preferences, nor did it produce consistent effects on any other behavioral measures. In contrast, elevating the relative expression of Homer2b versus Homer1 within mPFC, by overexpressing Homer2b or knocking down Homer1c, shifted the dose-response function for cocaine-conditioned reward to the left, without affecting cocaine locomotion or sensitization. Intriguingly, both these transgenic manipulations produced glutamate anomalies within the nucleus accumbens (NAC) of cocaine-naive animals that are reminiscent of those observed in cocaine experienced animals, including reduced basal extracellular glutamate content, reduced Homer1/2 and glutamate receptor expression, and augmented cocaine-elicited glutamate release. Together, these data provide novel evidence in support of opposing roles for constitutively expressed Homer1 and Homer2 isoforms in regulating mPFC glutamate transmission in vivo and support the hypothesis that cocaine-elicited increases in the relative amount of mPFC Homer2 versus Homer1 signaling produces abnormalities in NAC glutamate transmission that enhance vulnerability to cocaine reward.
Collapse
|
39
|
Bai X, Twaroski D, Bosnjak ZJ. Modeling anesthetic developmental neurotoxicity using human stem cells. Semin Cardiothorac Vasc Anesth 2013; 17:276-87. [PMID: 23859832 DOI: 10.1177/1089253213495923] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mounting preclinical evidence in rodents and nonhuman primates has demonstrated that prolonged exposure of developing animals to general anesthetics can induce widespread neuronal cell death followed by long-term memory and learning disabilities. In vitro experimental evidence from cultured neonatal animal neurons confirmed the in vivo findings. However, there is no direct clinical evidence of the detrimental effects of anesthetics in human fetuses, infants, or children. Development of an in vitro neurogenesis system using human stem cells has opened up avenues of research for advancing our understanding of human brain development and the issues relevant to anesthetic-induced developmental toxicity in human neuronal lineages. Recent studies from our group, as well as other groups, showed that isoflurane influences human neural stem cell proliferation and neurogenesis, whereas ketamine induces neuroapoptosis. Application of this high throughput in vitro stem cell neurogenesis approach is a major stride toward ensuring the safety of anesthetic agents in young children. This in vitro human model allows us to (1) screen the toxic effects of various anesthetics under controlled conditions during intense neuronal growth, (2) find the trigger for the anesthetic-induced catastrophic chain of toxic events, and (3) develop prevention strategies to avoid this toxic effect. In this article, we reviewed the current findings in anesthetic-induced neurotoxicity studies, specifically focusing on the in vitro human stem cell model.
Collapse
Affiliation(s)
- Xiaowen Bai
- 1Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | |
Collapse
|
40
|
Fatemi SH, Folsom TD, Kneeland RE, Yousefi MK, Liesch SB, Thuras PD. Impairment of fragile X mental retardation protein-metabotropic glutamate receptor 5 signaling and its downstream cognates ras-related C3 botulinum toxin substrate 1, amyloid beta A4 precursor protein, striatal-enriched protein tyrosine phosphatase, and homer 1, in autism: a postmortem study in cerebellar vermis and superior frontal cortex. Mol Autism 2013; 4:21. [PMID: 23803181 PMCID: PMC3702477 DOI: 10.1186/2040-2392-4-21] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 05/14/2013] [Indexed: 12/18/2022] Open
Abstract
Background Candidate genes associated with idiopathic forms of autism overlap with other disorders including fragile X syndrome. Our laboratory has previously shown reduction in fragile X mental retardation protein (FMRP) and increase in metabotropic glutamate receptor 5 (mGluR5) in cerebellar vermis and superior frontal cortex (BA9) of individuals with autism. Methods In the current study we have investigated expression of four targets of FMRP and mGluR5 signaling - homer 1, amyloid beta A4 precursor protein (APP), ras-related C3 botulinum toxin substrate 1 (RAC1), and striatal-enriched protein tyrosine phosphatase (STEP) - in the cerebellar vermis and superior frontal cortex (BA9) via SDS-PAGE and western blotting. Data were analyzed based on stratification with respect to age (children and adolescents vs. adults), anatomic region of the brain (BA9 vs. cerebellar vermis), and impact of medications (children and adolescents on medications (n = 4) vs. total children and adolescents (n = 12); adults on medications (n = 6) vs. total adults (n = 12)). Results There were significant increases in RAC1, APP 120 kDa and APP 80 kDa proteins in BA9 of children with autism vs. healthy controls. None of the same proteins were significantly affected in cerebellar vermis of children with autism. In BA9 of adults with autism there were significant increases in RAC1 and STEP 46 kDa and a significant decrease in homer 1 vs. controls. In the vermis of adult subjects with autism, RAC1 was significantly increased while APP 120, STEP 66 kDa, STEP 27 kDa, and homer 1 were significantly decreased when compared with healthy controls. No changes were observed in vermis of children with autism. There was a significant effect of anticonvulsant use on STEP 46 kDa/β-actin and a potential effect on homer 1/NSE, in BA9 of adults with autism. However, no other significant confound effects were observed in this study. Conclusions Our findings provide further evidence of abnormalities in FMRP and mGluR5 signaling partners in brains of individuals with autism and open the door to potential targeted treatments which could help ameliorate the symptoms of autism.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Obara I, Goulding SP, Hu JH, Klugmann M, Worley PF, Szumlinski KK. Nerve injury-induced changes in Homer/glutamate receptor signaling contribute to the development and maintenance of neuropathic pain. Pain 2013; 154:1932-1945. [PMID: 23685007 DOI: 10.1016/j.pain.2013.03.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 02/12/2013] [Accepted: 03/26/2013] [Indexed: 11/28/2022]
Abstract
While group 1 metabotropic glutamate receptors (mGluRs) and ionotropic N-methyl-d-aspartate (NMDA) receptors regulate nociception, the precise molecular mechanism(s) contributing to glutamate signaling in chronic pain remain unclear. Here we not only confirmed the key involvement of Homer proteins in neuropathic pain, but also distinguished between the functional roles for different Homer family members and isoforms. Chronic constriction injury (CCI) of the sciatic nerve induced long-lasting, time-dependent increases in the postsynaptic density expression of the constitutively expressed (CC) isoforms Homer1b/c and/or Homer2a/b in the spinal dorsal horn and supraspinal structures involved in nociception (prefrontal cortex, thalamus), that co-occurred with increases in their associated mGluRs, NR2 subunits of the NMDA receptor, and the activation of downstream kinases. Virus-mediated overexpression of Homer1c and Homer2b after spinal (intrathecal) virus injection exacerbated CCI-induced mechanical and cold hypersensitivity, however, Homer1 and Homer2 gene knockout (KO) mice displayed no changes in their neuropathic phenotype. In contrast, overexpression of the immediate early gene (IEG) Homer1a isoform reduced, while KO of Homer1a gene potentiated neuropathic pain hypersensitivity. Thus, nerve injury-induced increases in CC-Homers expression promote pain in pathological states, but IEG-Homer induction protects against both the development and maintenance of neuropathy. Additionally, exacerbated pain hypersensitivity in transgenic mice with reduced Homer binding to mGluR5 supports also an inhibitory role for Homer interactions with mGluR5 in mediating neuropathy. Such data indicate that nerve injury-induced changes in glutamate receptor/Homer signaling contribute in dynamic but distinct ways to neuropathic pain processing, which has relevance for the etiology of chronic pain symptoms and its treatment.
Collapse
Affiliation(s)
- Ilona Obara
- Department of Psychology and The Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, CA 93106-9660, USA School of Medicine, Pharmacy and Health, Durham University, Queens Campus, Stockton on Tees TS17 6BH, UK Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Translational Neuroscience Facility, School of Medical Sciences, UNSW Kensington Campus, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | | | | | |
Collapse
|
42
|
Esseltine JL, Willard MD, Wulur IH, Lajiness ME, Barber TD, Ferguson SSG. Somatic mutations in GRM1 in cancer alter metabotropic glutamate receptor 1 intracellular localization and signaling. Mol Pharmacol 2013; 83:770-80. [PMID: 23303475 DOI: 10.1124/mol.112.081695] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The activity of metabotropic glutamate receptors (mGluRs) is known to be altered as the consequence of neurodegenerative diseases such as Alzheimer, Parkinson, and Huntington disease. However, little attention has been paid to this receptor family's potential link with cancer. Recent reports indicate altered mGluR signaling in various tumor types, and several somatic mutations in mGluR1a in lung cancer were recently described. Group 1 mGluRs (mGluR1a and mGluR5) are coupled primarily to Gαq, leading to the activation of phospholipase C and to the formation of diacylglycerol and inositol 1,4,5-trisphosphate, leading to the release of Ca(2+) from intracellular stores and protein kinase C (PKC) activation. In the present study, we investigated the intracellular localization and G protein-dependent and -independent signaling of eight GRM1 (mGluR1a) somatic mutations. Two mutants found in close proximity to the glutamate binding domain and cysteine-rich region (R375G and G396V) show both decreased cell surface expression and basal inositol phosphate (IP) formation. However, R375G shows increased ERK1/2 activation in response to quisqualate stimulation. A mutant located directly in the glutamate binding site (A168V) shows increased quisqualate-induced IP formation and, similar to R375G, increased ERK1/2 activation. Additionally, a mutation in the G protein-coupled receptor kinase 2/PKC regulatory region (R696W) shows decreased ERK1/2 activation, whereas a mutation within the Homer binding region in the carboxyl-terminal tail (P1148L) does not alter the intracellular localization of the receptor, but it induces changes in cellular morphology and exhibits reduced ERK1/2 activation. Taken together, these results suggest that mGluR1a signaling in cancer is disrupted by somatic mutations with multiple downstream consequences.
Collapse
Affiliation(s)
- Jessica L Esseltine
- Molecular Brain Research Group, Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Bai X, Yan Y, Canfield S, Muravyeva MY, Kikuchi C, Zaja I, Corbett JA, Bosnjak ZJ. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg 2013; 116:869-80. [PMID: 23460563 DOI: 10.1213/ane.0b013e3182860fc9] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Growing evidence indicates that ketamine causes neurotoxicity in a variety of developing animal models, leading to a serious concern regarding the safety of pediatric anesthesia. However, if and how ketamine induces human neural cell toxicity is unknown. Recapitulation of neurogenesis from human embryonic stem cells (hESCs) in vitro allows investigation of the toxic effects of ketamine on neural stem cells (NSCs) and developing neurons, which is impossible to perform in humans. In the present study, we assessed the influence of ketamine on the hESC-derived NSCs and neurons. METHODS hESCs were directly differentiated into neurons via NSCs. NSCs and 2-week-old neurons were treated with varying doses of ketamine for different durations. NSC proliferation capacity was analyzed by Ki67 immunofluorescence staining and bromodeoxyuridine assay. Neuroapoptosis was analyzed by TUNEL staining and caspase 3 activity measurement. The mitochondria-related neuronal apoptosis pathway including mitochondrial membrane potential, cytochrome c distribution within cells, mitochondrial fission, and reactive oxygen species (ROS) production were also investigated. RESULTS Ketamine (100 µM) increased NSC proliferation after 6-hour exposure. However, significant neuronal apoptosis was only observed after 24 hours of ketamine treatment. In addition, ketamine decreased mitochondrial membrane potential and increased cytochrome c release from mitochondria into cytosol. Ketamine also enhanced mitochondrial fission as well as ROS production compared with no-treatment control. Importantly, Trolox, a ROS scavenger, significantly attenuated the increase of ketamine-induced ROS production and neuronal apoptosis. CONCLUSIONS These data for the first time demonstrate that (1) ketamine increases NSC proliferation and causes neuronal apoptosis; (2) mitochondria are involved in ketamine-induced neuronal toxicity, which can be prevented by Trolox; and (3) the stem cell-associated neurogenesis system may provide a simple and promising in vitro model for rapidly screening anesthetic neurotoxicity and studying the underlying mechanisms as well as prevention strategies to avoid this toxic effect.
Collapse
Affiliation(s)
- Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Li Y, Popko J, Krogh KA, Thayer SA. Epileptiform stimulus increases Homer 1a expression to modulate synapse number and activity in hippocampal cultures. J Neurophysiol 2012; 109:1494-504. [PMID: 23274309 DOI: 10.1152/jn.00580.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurons adapt to seizure activity structurally and functionally to attenuate hyperactive neural circuits. Homer proteins provide a scaffold in the postsynaptic density (PSD) by binding to ligands through an EVH1 domain and to other Homer proteins by a coiled-coil domain. The short Homer isoform 1a (H1a) has a ligand-binding domain but lacks a coiled-coil domain and thus acts in a dominant-negative manner to uncouple Homer scaffolds. Here, we show that treating rat hippocampal cultures with bicuculline and 4-aminopyridine (Bic+4-AP) evoked epileptiform activity and synchronized Ca(2+) spiking, measured with whole cell current-clamp and fura-2-based digital imaging; Bic+4-AP increased H1a mRNA through the activation of metabotropic glutamate receptor 5 (mGluR5). Treatment with Bic+4-AP for 4 h attenuated burst firing and induced synapse loss. Synaptic changes were measured using a confocal imaging-based assay that quantified clusters of PSD-95 fused to green fluorescent protein. Treatment with an mGluR5 antagonist blocked H1a expression, synapse loss, and burst attenuation. Overexpression of H1a inhibited burst firing similar to Bic+4-AP treatment. Furthermore, knockdown of H1a using a short hairpin RNA (shRNA) strategy reduced synapse loss and burst attenuation induced by Bic+4-AP treatment. Thus an epileptiform stimulus applied to hippocampal neurons in culture induced burst firing and H1a expression through the activation of mGluR5; a 4-h exposure to this stimulus resulted in synapse loss and burst attenuation. These results suggest that H1a expression functions in a negative-feedback manner to reduce network excitability by regulating the number of synapses.
Collapse
Affiliation(s)
- Yan Li
- Dept. of Pharmacology, Univ. of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
45
|
Scaffolding proteins of the post-synaptic density contribute to synaptic plasticity by regulating receptor localization and distribution: relevance for neuropsychiatric diseases. Neurochem Res 2012; 38:1-22. [PMID: 22991141 DOI: 10.1007/s11064-012-0886-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/16/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
Synaptic plasticity represents the long lasting activity-related strengthening or weakening of synaptic transmission, whose well-characterized types are the long term potentiation and depression. Despite this classical definition, however, the molecular mechanisms by which synaptic plasticity may occur appear to be extremely complex and various. The post-synaptic density (PSD) of glutamatergic synapses is a major site for synaptic plasticity processes and alterations of PSD members have been recently implicated in neuropsychiatric diseases where an impairment of synaptic plasticity has also been reported. Among PSD members, scaffolding proteins have been demonstrated to bridge surface receptors with their intracellular effectors and to regulate receptors distribution and localization both at surface membranes and within the PSD. This review will focus on the molecular physiology and pathophysiology of synaptic plasticity processes, which are tuned by scaffolding PSD proteins and their close related partners, through the modulation of receptor localization and distribution at post-synaptic sites. We suggest that, by regulating both the compartmentalization of receptors along surface membrane and their degradation as well as by modulating receptor trafficking into the PSD, postsynaptic scaffolding proteins may contribute to form distinct signaling micro-domains, whose efficacy in transmitting synaptic signals depends on the dynamic stability of the scaffold, which in turn is provided by relative amounts and post-translational modifications of scaffolding members. The putative relevance for neuropsychiatric diseases and possible pathophysiological mechanisms are discussed in the last part of this work.
Collapse
|
46
|
Moutin E, Raynaud F, Roger J, Pellegrino E, Homburger V, Bertaso F, Ollendorff V, Bockaert J, Fagni L, Perroy J. Dynamic remodeling of scaffold interactions in dendritic spines controls synaptic excitability. ACTA ACUST UNITED AC 2012; 198:251-63. [PMID: 22801779 PMCID: PMC3410417 DOI: 10.1083/jcb.201110101] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Synaptic activity–dependent remodeling of the glutamate receptor scaffold complex generates a negative feedback loop that limits further NMDA receptor activation. Scaffolding proteins interact with membrane receptors to control signaling pathways and cellular functions. However, the dynamics and specific roles of interactions between different components of scaffold complexes are poorly understood because of the dearth of methods available to monitor binding interactions. Using a unique combination of single-cell bioluminescence resonance energy transfer imaging in living neurons and electrophysiological recordings, in this paper, we depict the role of glutamate receptor scaffold complex remodeling in space and time to control synaptic transmission. Despite a broad colocalization of the proteins in neurons, we show that spine-confined assembly/disassembly of this scaffold complex, physiologically triggered by sustained activation of synaptic NMDA (N-methyl-d-aspartate) receptors, induces physical association between ionotropic (NMDA) and metabotropic (mGlu5a) synaptic glutamate receptors. This physical interaction results in an mGlu5a receptor–mediated inhibition of NMDA currents, providing an activity-dependent negative feedback loop on NMDA receptor activity. Such protein scaffold remodeling represents a form of homeostatic control of synaptic excitability.
Collapse
Affiliation(s)
- Enora Moutin
- Centre national de la recherche scientifique, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, Cedex 16, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Li Y, Krogh KA, Thayer SA. Epileptic stimulus increases Homer 1a expression to modulate endocannabinoid signaling in cultured hippocampal neurons. Neuropharmacology 2012; 63:1140-9. [PMID: 22814532 DOI: 10.1016/j.neuropharm.2012.07.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 06/12/2012] [Accepted: 07/03/2012] [Indexed: 02/06/2023]
Abstract
Endocannabinoid (eCB) signaling serves as an on-demand neuroprotective system. eCBs are produced postsynaptically in response to depolarization or activation of metabotropic glutamate receptors (mGluRs) and act on presynaptic cannabinoid receptor-1 to suppress synaptic transmission. Here, we examined the effects of epileptiform activity on these two forms of eCB signaling in hippocampal cultures. Treatment with bicuculline and 4-aminopyridine (Bic + 4-AP), which induced burst firing, inhibited metabotropic-induced suppression of excitation (MSE) and prolonged the duration of depolarization-induced suppression of excitation (DSE). The Homer family of proteins provides a scaffold for signaling molecules including mGluRs. It is known that seizures induce the expression of the short Homer isoform 1a (H1a) that acts in a dominant negative manner to uncouple Homer scaffolds. Bic + 4-AP treatment increased H1a mRNA. A group I mGluR antagonist blocked the Bic + 4-AP-evoked increase in burst firing, the increase in H1a expression, and the inhibition of MSE. Bic + 4-AP treatment reduced mGluR-mediated Ca(2+) mobilization from inositol trisphosphate-sensitive stores relative to untreated cells. Expression of H1a, but not a mutant form that cannot bind Homer ligands, mimicked Bic + 4-AP inhibition of MSE and mGluR-mediated Ca(2+) mobilization. In cells expressing shRNA targeted to Homer 1 mRNA, Bic + 4-AP did not affect mGluR-mediated Ca(2+) release. Furthermore, knockdown of H1a prevented the inhibition of MSE induced by Bic + 4-AP. Thus, an epileptic stimulus increased H1a expression, which subsequently uncoupled mGluR-mediated eCB production. These results indicate that seizure activity modulates eCB-mediated synaptic plasticity, suggesting a changing role for the eCB system following exposure to aberrant patterns of excitatory synaptic activity.
Collapse
Affiliation(s)
- Yan Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
48
|
de Bartolomeis A, Tomasetti C. Calcium-Dependent Networks in Dopamine–Glutamate Interaction: The Role of Postsynaptic Scaffolding Proteins. Mol Neurobiol 2012; 46:275-96. [DOI: 10.1007/s12035-012-8293-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 06/21/2012] [Indexed: 01/11/2023]
|
49
|
Beqollari D, Kammermeier PJ. The interaction between mGluR1 and the calcium channel Cav₂.₁ preserves coupling in the presence of long Homer proteins. Neuropharmacology 2012; 66:302-10. [PMID: 22659088 DOI: 10.1016/j.neuropharm.2012.05.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 05/18/2012] [Accepted: 05/21/2012] [Indexed: 11/29/2022]
Abstract
Group I metabotropic glutamate receptors (mGluR1 and 5) are G protein coupled receptors that regulate neuronal activity in a number of ways. Some of the most well studied functions of group I mGluRs, such as initiation of multiple forms of mGluR-dependent long-term depression, require receptor localization near the post-synaptic density (PSD). This localization is in turn dependent on the Homer family of scaffolding proteins which bind to a small motif on the distal C-termini of mGluR1 and 5, localize the receptors near the PSD, strengthen coupling to post-synaptic effectors and simultaneously uncouple the mGluRs from extra-synaptic effectors such as voltage dependent ion channels. Here the selectivity of this uncoupling process was examined by testing the ability of Homer-2b to uncouple mGluR1 from multiple voltage dependent calcium channels including Ca(V2.2) (N-type), Ca(V3.2) (T-type), and Ca(V2.1) (P/Q-type) expressed in rat sympathetic neurons from the superior cervical ganglion (SCG). Of these, only the mGluR1-Ca(V2.1) modulatory pathway was insensitive to Homer-2b expression. Uncoupling from this channel was achieved by co-expression of an mGluR1 C-terminal protein designed to disrupt a previously described direct interaction between these two proteins, suggesting that this interaction allows incorporation of Ca(V2.1) into the mGluR1/Homer signaling complex, thereby preserving modulation in the presence of scaffolding Homer proteins. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- Donald Beqollari
- University of Rochester Medical Center, Department of Pharmacology and Physiology, Rochester, NY 14642, USA
| | | |
Collapse
|
50
|
Ting JT, Peça J, Feng G. Functional consequences of mutations in postsynaptic scaffolding proteins and relevance to psychiatric disorders. Annu Rev Neurosci 2012; 35:49-71. [PMID: 22540979 DOI: 10.1146/annurev-neuro-062111-150442] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Functional studies on postsynaptic scaffolding proteins at excitatory synapses have revealed a plethora of important roles for synaptic structure and function. In addition, a convergence of recent in vivo functional evidence together with human genetics data strongly suggest that mutations in a variety of these postsynaptic scaffolding proteins may contribute to the etiology of diverse human psychiatric disorders such as schizophrenia, autism spectrum disorders, and obsessive-compulsive spectrum disorders. Here we review the most recent evidence for several key postsynaptic scaffolding protein families and explore how mouse genetics and human genetics have intersected to advance our knowledge concerning the contributions of these important players to complex brain function and dysfunction.
Collapse
Affiliation(s)
- Jonathan T Ting
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | | | |
Collapse
|