1
|
Kim HJ, Lee S, Kim GH, Sung K, Yoo T, Pyo JH, Jo HJ, Lee S, Lee HY, Jung JH, Lee KJ, Kim JH. GluN2B-mediated regulation of silent synapses for receptor specification and addiction memory. Exp Mol Med 2025; 57:436-449. [PMID: 39930130 PMCID: PMC11873126 DOI: 10.1038/s12276-025-01399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 03/04/2025] Open
Abstract
Psychostimulants, including cocaine, elicit stereotyped, addictive behaviors. The reemergence of silent synapses containing only NMDA-type glutamate receptors is a critical mediator of addiction memory and seeking behaviors. Despite the predominant abundance of GluN2B-containing NMDA-type glutamate receptors in silent synapses, their operational mechanisms are not fully understood. Here, using conditional depletion/deletion of GluN2B in D1-expressing accumbal medium spiny neurons, we examined the synaptic and behavioral actions that silent synapses incur after repeated exposure to cocaine. GluN2B ablation reduces the proportion of silent synapses, but some of them can persist by substitution with GluN2C, which drives the aberrantly facilitated synaptic incorporation of calcium-impermeable AMPA-type glutamate receptors (AMPARs). The resulting precocious maturation of silent synapses impairs addiction memory but increases locomotor activity, both of which can be normalized by the blockade of calcium-impermeable AMPAR trafficking. Collectively, GluN2B supports the competence of cocaine-induced silent synapses to specify the subunit composition of AMPARs and thereby the expression of addiction memory and related behaviors.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sangjun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Gyu Hyun Kim
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kibong Sung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Taesik Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jung Hyun Pyo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hee-Jung Jo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sanghyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Young Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jung Hoon Jung
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Kea Joo Lee
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
2
|
Mould AW, Wright DJ, Bornemann KD, Hengerer B, Pinnock R, Drydale E, Bancroft J, Hall NAL, von Delft A, Brennan PE, Harrison PJ, Haerty W, Tunbridge EM. Identification and characterization of human KALRN mRNA and Kalirin protein isoforms. Cereb Cortex 2024; 34:bhae470. [PMID: 39656879 DOI: 10.1093/cercor/bhae470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Kalirin is a multidomain protein with important roles in neurite outgrowth, and synaptic spine formation and remodeling. Genetic and pathophysiological links with various neuropsychiatric disorders associated with synaptic dysfunction and cognitive impairment have sparked interest in its potential as a pharmacological target. Multiple Kalirin proteoforms are detected in the adult human brain, yet we know little about the diversity of the transcripts that encode them or their tissue profiles. Here, we characterized full-length KALRN transcripts expressed in the adult human frontal lobe and hippocampus using rapid amplification of complementary DNA (cDNA) ends and nanopore long-read sequencing. For comparison with non-neural tissue, we also analyzed KALRN transcripts in the aorta. Multiple novel isoforms were identified and were largely similar between the two brain regions analyzed. Alternative splicing in the brain results in preferential inclusion of exon 37, which encodes 32 amino acids upstream of the second guanine nucleotide exchange factor (GEF) domain. Structural modeling predicts that a subset of these amino acids forms a conserved alpha helix. Although deletion of these amino acids had little effect on GEF activity, it did alter Kalirin-induced neurite outgrowth suggesting that this brain-enriched splicing event may be important for neural function. These data indicate that alternative splicing is potentially important for regulating Kalirin actions in the human brain.
Collapse
Affiliation(s)
- Arne W Mould
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, United Kingdom
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, United Kingdom
| | - David J Wright
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, United Kingdom
| | - Klaus D Bornemann
- Boehringer Ingelheim Pharma GmbH & Co. KG, 65 Birkendorfer Straße, 88397, Biberach an der Riß, Germany
| | - Bastian Hengerer
- Boehringer Ingelheim Pharma GmbH & Co. KG, 65 Birkendorfer Straße, 88397, Biberach an der Riß, Germany
| | - Rob Pinnock
- Biogen Idec Ltd, 5 Roxborough Way, Maidenhead SL6 3UD, United Kingdom
| | - Edward Drydale
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - James Bancroft
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Nicola A L Hall
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, United Kingdom
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, United Kingdom
| | - Annette von Delft
- Centre for Medicines Discovery, NDM Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, United Kingdom
| | - Paul E Brennan
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, NDM Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, United Kingdom
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, United Kingdom
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, United Kingdom
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, United Kingdom
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, United Kingdom
- Boehringer Ingelheim Pharma GmbH & Co. KG, 65 Birkendorfer Straße, 88397, Biberach an der Riß, Germany
| |
Collapse
|
3
|
Powis G, Meuillet EJ, Indarte M, Booher G, Kirkpatrick L. Pleckstrin Homology [PH] domain, structure, mechanism, and contribution to human disease. Biomed Pharmacother 2023; 165:115024. [PMID: 37399719 DOI: 10.1016/j.biopha.2023.115024] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023] Open
Abstract
The pleckstrin homology [PH] domain is a structural fold found in more than 250 proteins making it the 11th most common domain in the human proteome. 25% of family members have more than one PH domain and some PH domains are split by one, or several other, protein domains although still folding to give functioning PH domains. We review mechanisms of PH domain activity, the role PH domain mutation plays in human disease including cancer, hyperproliferation, neurodegeneration, inflammation, and infection, and discuss pharmacotherapeutic approaches to regulate PH domain activity for the treatment of human disease. Almost half PH domain family members bind phosphatidylinositols [PIs] that attach the host protein to cell membranes where they interact with other membrane proteins to give signaling complexes or cytoskeleton scaffold platforms. A PH domain in its native state may fold over other protein domains thereby preventing substrate access to a catalytic site or binding with other proteins. The resulting autoinhibition can be released by PI binding to the PH domain, or by protein phosphorylation thus providing fine tuning of the cellular control of PH domain protein activity. For many years the PH domain was thought to be undruggable until high-resolution structures of human PH domains allowed structure-based design of novel inhibitors that selectively bind the PH domain. Allosteric inhibitors of the Akt1 PH domain have already been tested in cancer patients and for proteus syndrome, with several other PH domain inhibitors in preclinical development for treatment of other human diseases.
Collapse
Affiliation(s)
- Garth Powis
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA.
| | | | - Martin Indarte
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| | - Garrett Booher
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| | - Lynn Kirkpatrick
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| |
Collapse
|
4
|
Wu Q, Cai C, Ying X, Zheng Y, Yu J, Gu X, Tu W, Lou X, Yang G, Li M, Jiang S. Electroacupuncture inhibits dendritic spine remodeling through the srGAP3-Rac1 signaling pathway in rats with SNL. Biol Res 2023; 56:26. [PMID: 37211600 DOI: 10.1186/s40659-023-00439-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/10/2023] [Indexed: 05/23/2023] Open
Abstract
Previous studies have shown that peripheral nerve injury can lead to abnormal dendritic spine remodeling in spinal dorsal horn neurons. Inhibition of abnormal dendritic spine remodeling can relieve neuropathic pain. Electroacupuncture (EA) has a beneficial effect on the treatment of neuropathic pain, but the specific mechanism remains unclear. Evidence has shown that slit-robo GTPase activating protein 3 (srGAP3) and Rho GTPase (Rac1) play very important roles in dendritic spine remodeling. Here, we used srGAP3 siRNA and Rac1 activator CN04 to confirm the relationship between SrGAP3 and Rac1 and their roles in improving neuropathic pain with EA. Spinal nerve ligation (SNL) was used as the experimental model, and thermal withdrawal latency (TWL), mechanical withdrawal threshold (MWT), Western blotting, immunohistochemistry and Golgi-Cox staining were used to examine changes in behavioral performance, protein expression and dendritic spines. More dendritic spines and higher expression levels of srGAP3 were found in the initial phase of neuropathic pain. During the maintenance phase, dendritic spines were more mature, which was consistent with lower expression levels of srGAP3 and higher expression levels of Rac1-GTP. EA during the maintenance phase reduced the density and maturity of dendritic spines of rats with SNL, increased the levels of srGAP3 and reduced the levels of Rac1-GTP, while srGAP3 siRNA and CN04 reversed the therapeutic effects of EA. These results suggest that dendritic spines have different manifestations in different stages of neuropathic pain and that EA may inhibit the abnormal dendritic spine remodeling by regulating the srGAP3/Rac1 signaling pathway to alleviate neuropathic pain.
Collapse
Affiliation(s)
- Qiaoyun Wu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Chenchen Cai
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Xinwang Ying
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Yujun Zheng
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Jiaying Yu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Xiaoxue Gu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Wenzhan Tu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Xinfa Lou
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Guanhu Yang
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Ming Li
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China
| | - Songhe Jiang
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 268 Xue Yuan Xi Road, Wenzhou, 325027, Zhejiang, People's Republic of China.
- Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- The Wenzhou Key Laboratory for Rehabilitation Research, The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, Wenzhou, China.
| |
Collapse
|
5
|
Ru Q, Wang Y, Zhou E, Chen L, Wu Y. The potential therapeutic roles of Rho GTPases in substance dependence. Front Mol Neurosci 2023; 16:1125277. [PMID: 37063367 PMCID: PMC10097952 DOI: 10.3389/fnmol.2023.1125277] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Rho GTPases family are considered to be molecular switches that regulate various cellular processes, including cytoskeleton remodeling, cell polarity, synaptic development and maintenance. Accumulating evidence shows that Rho GTPases are involved in neuronal development and brain diseases, including substance dependence. However, the functions of Rho GTPases in substance dependence are divergent and cerebral nuclei-dependent. Thereby, comprehensive integration of their roles and correlated mechanisms are urgently needed. In this review, the molecular functions and regulatory mechanisms of Rho GTPases and their regulators such as GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs) in substance dependence have been reviewed, and this is of great significance for understanding their spatiotemporal roles in addictions induced by different addictive substances and in different stages of substance dependence.
Collapse
Affiliation(s)
| | | | | | - Lin Chen
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| | - Yuxiang Wu
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| |
Collapse
|
6
|
Parnell E, Voorn RA, Martin-de-Saavedra MD, Loizzo DD, Dos Santos M, Penzes P. A developmental delay linked missense mutation in Kalirin-7 disrupts protein function and neuronal morphology. Front Mol Neurosci 2022; 15:994513. [PMID: 36533124 PMCID: PMC9751355 DOI: 10.3389/fnmol.2022.994513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/28/2022] [Indexed: 07/30/2023] Open
Abstract
The Rac1 guanine exchange factor Kalirin-7 is a key regulator of dendritic spine morphology, LTP and dendritic arborization. Kalirin-7 dysfunction and genetic variation has been extensively linked to various neurodevelopmental and neurodegenerative disorders. Here we characterize a Kalirin-7 missense mutation, glu1577lys (E1577K), identified in a patient with severe developmental delay. The E1577K point mutation is located within the catalytic domain of Kalirin-7, and results in a robust reduction in Kalirin-7 Rac1 Guanosine exchange factor activity. In contrast to wild type Kalirin-7, the E1577K mutant failed to drive dendritic arborization, spine density, NMDAr targeting to, and activity within, spines. Together these results indicate that reduced Rac1-GEF activity as result of E1577K mutation impairs neuroarchitecture, connectivity and NMDAr activity, and is a likely contributor to impaired neurodevelopment in a patient with developmental delay.
Collapse
Affiliation(s)
- Euan Parnell
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Roos A. Voorn
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - M. Dolores Martin-de-Saavedra
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Instituto Universitario de Investigación en Neuroquímica, Complutense University of Madrid, Madrid, Spain
| | - Daniel D. Loizzo
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Marc Dos Santos
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Peter Penzes
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Centre for Autism and Neurodevelopment, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
7
|
Chronic Pain after Bone Fracture: Current Insights into Molecular Mechanisms and Therapeutic Strategies. Brain Sci 2022; 12:brainsci12081056. [PMID: 36009119 PMCID: PMC9406150 DOI: 10.3390/brainsci12081056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/20/2022] [Accepted: 08/06/2022] [Indexed: 12/12/2022] Open
Abstract
Bone fracture following traumatic injury or due to osteoporosis is characterized by severe pain and motor impairment and is a major cause of global mortality and disability. Fracture pain often originates from mechanical distortion of somatosensory nerve terminals innervating bones and muscles and is maintained by central sensitization. Chronic fracture pain (CFP) after orthopedic repairs is considered one of the most critical contributors to interference with the physical rehabilitation and musculoskeletal functional recovery. Analgesics available for CFP in clinics not only have poor curative potency but also have considerable side effects; therefore, it is important to further explore the pathogenesis of CFP and identify safe and effective therapies. The typical physiopathological characteristics of CFP are a neuroinflammatory response and excitatory synaptic plasticity, but the specific molecular mechanisms involved remain poorly elucidated. Recent progress has deepened our understanding of the emerging properties of chemokine production, proinflammatory mediator secretion, caspase activation, neurotransmitter release, and neuron-glia interaction in initiating and sustaining synaptogenesis, synaptic strength, and signal transduction in central pain sensitization, indicating the possibility of targeting neuroinflammation to prevent and treat CFP. This review summarizes current literature on the excitatory synaptic plasticity, microgliosis, and microglial activation-associated signaling molecules and discusses the unconventional modulation of caspases and stimulator of interferon genes (STING) in the pathophysiology of CFP. We also review the mechanisms of action of analgesics in the clinic and their side effects as well as promising therapeutic candidates (e.g., specialized pro-resolving mediators, a caspase-6 inhibitor, and a STING agonist) for pain relief by the attenuation of neuroinflammation with the aim of better managing patients undergoing CFP in the clinical setting.
Collapse
|
8
|
Viard J, Loe-Mie Y, Daudin R, Khelfaoui M, Plancon C, Boland A, Tejedor F, Huganir RL, Kim E, Kinoshita M, Liu G, Haucke V, Moncion T, Yu E, Hindie V, Bléhaut H, Mircher C, Herault Y, Deleuze JF, Rain JC, Simonneau M, Lepagnol-Bestel AM. Chr21 protein-protein interactions: enrichment in proteins involved in intellectual disability, autism, and late-onset Alzheimer's disease. Life Sci Alliance 2022; 5:e202101205. [PMID: 35914814 PMCID: PMC9348576 DOI: 10.26508/lsa.202101205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Down syndrome (DS) is caused by human chromosome 21 (HSA21) trisomy. It is characterized by a poorly understood intellectual disability (ID). We studied two mouse models of DS, one with an extra copy of the <i>Dyrk1A</i> gene (189N3) and the other with an extra copy of the mouse Chr16 syntenic region (Dp(16)1Yey). RNA-seq analysis of the transcripts deregulated in the embryonic hippocampus revealed an enrichment in genes associated with chromatin for the 189N3 model, and synapses for the Dp(16)1Yey model. A large-scale yeast two-hybrid screen (82 different screens, including 72 HSA21 baits and 10 rebounds) of a human brain library containing at least 10<sup>7</sup> independent fragments identified 1,949 novel protein-protein interactions. The direct interactors of HSA21 baits and rebounds were significantly enriched in ID-related genes (<i>P</i>-value < 2.29 × 10<sup>-8</sup>). Proximity ligation assays showed that some of the proteins encoded by HSA21 were located at the dendritic spine postsynaptic density, in a protein network at the dendritic spine postsynapse. We located HSA21 DYRK1A and DSCAM, mutations of which increase the risk of autism spectrum disorder (ASD) 20-fold, in this postsynaptic network. We found that an intracellular domain of DSCAM bound either DLGs, which are multimeric scaffolds comprising receptors, ion channels and associated signaling proteins, or DYRK1A. The DYRK1A-DSCAM interaction domain is conserved in <i>Drosophila</i> and humans. The postsynaptic network was found to be enriched in proteins associated with ARC-related synaptic plasticity, ASD, and late-onset Alzheimer's disease. These results highlight links between DS and brain diseases with a complex genetic basis.
Collapse
Affiliation(s)
- Julia Viard
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Yann Loe-Mie
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Rachel Daudin
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Malik Khelfaoui
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Christine Plancon
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Anne Boland
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Francisco Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Universidad Miguel Hernandez-Campus de San Juan, San Juan, Spain
| | - Richard L Huganir
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Institut für Molekulare Pharmakologie (FMP) and Freie Universität Berlin, Berlin, Germany
| | | | - Eugene Yu
- Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, State University of New York at Buffalo, Buffalo, NY, USA
| | | | | | | | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
- PHENOMIN, Institut Clinique de la Souris, ICS, GIE CERBM, CNRS, INSERM, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Jean-François Deleuze
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | | | - Michel Simonneau
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, CentraleSupélec, LuMIn, Gif sur Yvette, France
- Department of Biology, Ecole Normale Supérieure Paris-Saclay Université Paris-Saclay, Gif sur Yvette, France
| | | |
Collapse
|
9
|
Abstract
The cognitive dysfunction experienced by patients with schizophrenia represents a major unmet clinical need. We believe that enhancing synaptic function and plasticity by targeting kalirin may provide a novel means to remediate these symptoms. Karilin (a protein encoded by the KALRN gene) has multiple functional domains, including two Dbl homology (DH) guanine exchange factor (GEF) domains, which act to enhance the activity of the Rho family guanosine triphosphate (GTP)-ases. Here, we provide an overview of kalirin's roles in brain function and its therapeutic potential in schizophrenia. We outline how it mediates diverse effects via a suite of distinct isoforms that couple to members of the Rho GTPase family to regulate synapse formation and stabilisation, and how genomic and post-mortem data implicate it in schizophrenia. We then review the current state of knowledge about the influence of kalirin on brain function at a systems level, based largely on evidence from transgenic mouse models, which support its proposed role in regulating dendritic spine function and plasticity. We demonstrate that, whilst the GTPases are classically considered to be 'undruggable', targeting kalirin and other Rho GEFs provides a means to indirectly modulate their activity. Finally, we integrate across the information presented to assess the therapeutic potential of kalirin for schizophrenia and highlight the key outstanding questions required to advance it in this capacity; namely, the need for more information about the diversity and function of its isoforms, how these change across neurodevelopment, and how they affect brain function in vivo.
Collapse
|
10
|
Scala M, Nishikawa M, Nagata KI, Striano P. Pathophysiological Mechanisms in Neurodevelopmental Disorders Caused by Rac GTPases Dysregulation: What's behind Neuro-RACopathies. Cells 2021; 10:3395. [PMID: 34943902 PMCID: PMC8699292 DOI: 10.3390/cells10123395] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Rho family guanosine triphosphatases (GTPases) regulate cellular signaling and cytoskeletal dynamics, playing a pivotal role in cell adhesion, migration, and cell cycle progression. The Rac subfamily of Rho GTPases consists of three highly homologous proteins, Rac 1-3. The proper function of Rac1 and Rac3, and their correct interaction with guanine nucleotide-exchange factors (GEFs) and GTPase-activating proteins (GAPs) are crucial for neural development. Pathogenic variants affecting these delicate biological processes are implicated in different medical conditions in humans, primarily neurodevelopmental disorders (NDDs). In addition to a direct deleterious effect produced by genetic variants in the RAC genes, a dysregulated GTPase activity resulting from an abnormal function of GEFs and GAPs has been involved in the pathogenesis of distinctive emerging conditions. In this study, we reviewed the current pertinent literature on Rac-related disorders with a primary neurological involvement, providing an overview of the current knowledge on the pathophysiological mechanisms involved in the neuro-RACopathies.
Collapse
Affiliation(s)
- Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy;
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Masashi Nishikawa
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan; (M.N.); (K.-i.N.)
| | - Koh-ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan; (M.N.); (K.-i.N.)
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Nagoya 466-8550, Japan
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy;
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
11
|
Zhu Y, Qu Y, Zhang J, Hou J, Fang J, Shen J, Xu C, Huang M, Qiao H, An S. Phencynonate hydrochloride exerts antidepressant effects by regulating the dendritic spine density and altering glutamate receptor expression. Behav Pharmacol 2021; 32:660-672. [PMID: 34751176 DOI: 10.1097/fbp.0000000000000660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Phencynonate hydrochloride (PCH) is a drug that crosses the blood-brain barrier. Cellular experiments confirmed that PCH protects against glutamate toxicity and causes only weak central inhibition and limited side effects. As shown in our previous studies, PCH alleviates depression-like behaviours induced by chronic unpredictable mild stress (CUMS). Here we administered PCH at three different doses (4, 8 and 16 mg/kg) to male rats for two continuous days after CUMS and conducted behavioural tests to assess the dose-dependent antidepressant effects of PCH and its effects on the neuroplasticity in the hippocampus and medial prefrontal cortex (mPFC). Meanwhile, we measured the spine density and expression of related proteins to illustrate the mechanism of PCH. PCH treatment (8 mg/kg) significantly alleviated depression-like behaviours induced by CUMS. All doses of PCH treatment reversed the spine loss in prelimbic and CA3 regions induced by CUMS. Kalirin-7 expression was decreased in the hippocampus and mPFC of the CUMS group. The expression of the NR1 and NR2B subunits in the hippocampus, and NR2B in mPFC are increased by CUMS. PCH treatment (8 and 16 mg/kg) reversed all of these changes of Kalirin-7 in PFC and hippocampus, as well as NR1 and NR2B expression in the hippocampus. PCH is expected to be developed as a new type of rapid antidepressant. Its antidepressant effect may be closely related to the modulation of dendritic spine density in the prelimbic and CA3 regions and the regulation of Kalilin-7 and N-methyl-D-aspartic acid receptor levels in the hippocampus.
Collapse
Affiliation(s)
- Yingqi Zhu
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Yishan Qu
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jing Zhang
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jun Hou
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jie Fang
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jingxuan Shen
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Chang Xu
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Minyi Huang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, Hunan, China
| | - Hui Qiao
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Shucheng An
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| |
Collapse
|
12
|
Cheng J, Scala F, Blanco FA, Niu S, Firozi K, Keehan L, Mulherkar S, Froudarakis E, Li L, Duman JG, Jiang X, Tolias KF. The Rac-GEF Tiam1 Promotes Dendrite and Synapse Stabilization of Dentate Granule Cells and Restricts Hippocampal-Dependent Memory Functions. J Neurosci 2021; 41:1191-1206. [PMID: 33328293 PMCID: PMC7888217 DOI: 10.1523/jneurosci.3271-17.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
The dentate gyrus (DG) controls information flow into the hippocampus and is critical for learning, memory, pattern separation, and spatial coding, while DG dysfunction is associated with neuropsychiatric disorders. Despite its importance, the molecular mechanisms regulating DG neural circuit assembly and function remain unclear. Here, we identify the Rac-GEF Tiam1 as an important regulator of DG development and associated memory processes. In the hippocampus, Tiam1 is predominantly expressed in the DG throughout life. Global deletion of Tiam1 in male mice results in DG granule cells with simplified dendritic arbors, reduced dendritic spine density, and diminished excitatory synaptic transmission. Notably, DG granule cell dendrites and synapses develop normally in Tiam1 KO mice, resembling WT mice at postnatal day 21 (P21), but fail to stabilize, leading to dendrite and synapse loss by P42. These results indicate that Tiam1 promotes DG granule cell dendrite and synapse stabilization late in development. Tiam1 loss also increases the survival, but not the production, of adult-born DG granule cells, possibly because of greater circuit integration as a result of decreased competition with mature granule cells for synaptic inputs. Strikingly, both male and female mice lacking Tiam1 exhibit enhanced contextual fear memory and context discrimination. Together, these results suggest that Tiam1 is a key regulator of DG granule cell stabilization and function within hippocampal circuits. Moreover, based on the enhanced memory phenotype of Tiam1 KO mice, Tiam1 may be a potential target for the treatment of disorders involving memory impairments.SIGNIFICANCE STATEMENT The dentate gyrus (DG) is important for learning, memory, pattern separation, and spatial navigation, and its dysfunction is associated with neuropsychiatric disorders. However, the molecular mechanisms controlling DG formation and function remain elusive. By characterizing mice lacking the Rac-GEF Tiam1, we demonstrate that Tiam1 promotes the stabilization of DG granule cell dendritic arbors, spines, and synapses, whereas it restricts the survival of adult-born DG granule cells, which compete with mature granule cells for synaptic integration. Notably, mice lacking Tiam1 also exhibit enhanced contextual fear memory and context discrimination. These findings establish Tiam1 as an essential regulator of DG granule cell development, and identify it as a possible therapeutic target for memory enhancement.
Collapse
Affiliation(s)
- Jinxuan Cheng
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Federico Scala
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Francisco A Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, Texas 77030
| | - Sanyong Niu
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Laura Keehan
- Department of Biosciences, Rice University, Houston, Texas 77005
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | | | - Lingyong Li
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Joseph G Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Kimberley F Tolias
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
13
|
Zhang L, Wang Z, Song C, Liu H, Li Y, Li J, Yu Y, Wang G, Cui W. Spinal NR2B phosphorylation at Tyr1472 regulates IRE(-)DMT1-mediated iron accumulation and spine morphogenesis via kalirin-7 in tibial fracture-associated postoperative pain after orthopedic surgery in female mice. Reg Anesth Pain Med 2020; 46:363-373. [PMID: 33443215 DOI: 10.1136/rapm-2020-101883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Prolonged postoperative pain is a major concern and occurs more frequently in women, but mechanisms remain elusive. NR2B-containging N-methyl-d-aspartate (NMDA) receptor is a key component of nociception transduction. Divalent metal transporter 1 (DMT1)-mediated iron overload involves NMDA-induced neurotoxicity in males. Kalirin-7 is vital in synaptic plasticity underlying pathological pain in males. Herein, the requirement for kalirin-7 in NR2B phosphorylation-dependent iron accumulation and spine plasticity in postoperative pain after tibial fracture in female mice has been examined. METHODS Pain-related behavior, spinal NR2B phosphorylation at Tyr1472, kalirin-7 expression, DMT1 with/without iron-responsive element (IRE (+) DMT1 and IRE (-) DMT1) level, iron concentration and spine morphology were assessed in females. NR2B antagonist Ro25-6981, iron chelator deferoxamine and kalirin-7 knockdown by short hairpin RNA were employed to assess the potential cascade. RESULTS Tibial fracture initiates long-term allodynia lasting at least 21 days postoperatively, and upregulates spinal NR2B phosphorylation, kalirin-7 and IRE (-) DMT1 expression, iron overload and spine density. Ro25-6981 reduces postoperative mechanical and cold allodynia, spinal NR2B phosphorylation, kalirin-7 level and IRE (-) DMT1-mediated iron overload. Kalirin-7 knockdown impairs fracture-associated allodynia, IRE (-) DMT1-mediated iron overload and spine plasticity. Deferoxamine also attenuates behavioral allodynia and spine plasticity. Spinal NMDA application elicits NR2B-dependent mechanical allodynia and iron overload, which is reversed by kalirin-7 knockdown or coadministration of deferoxamine. CONCLUSION Spinal NR2B phosphorylation at Tyr1472 upregulates kalirin-7 expression to facilitate IRE (-) DMT1-mediated iron accumulation and spine morphogenesis in the development of fracture-associated postoperative pain in female mice.
Collapse
Affiliation(s)
- Linlin Zhang
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhen Wang
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chengcheng Song
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Haoyu Liu
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yize Li
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Li
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yonghao Yu
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guolin Wang
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Cui
- Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Stress-Sensitive Protein Rac1 and Its Involvement in Neurodevelopmental Disorders. Neural Plast 2020; 2020:8894372. [PMID: 33299404 PMCID: PMC7707960 DOI: 10.1155/2020/8894372] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/01/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Ras-related C3 botulinum toxin substrate 1 (Rac1) is a small GTPase that is well known for its sensitivity to the environmental stress of a cell or an organism. It senses the external signals which are transmitted from membrane-bound receptors and induces downstream signaling cascades to exert its physiological functions. Rac1 is an important regulator of a variety of cellular processes, such as cytoskeletal organization, generation of oxidative products, and gene expression. In particular, Rac1 has a significant influence on certain brain functions like neuronal migration, synaptic plasticity, and memory formation via regulation of actin dynamics in neurons. Abnormal Rac1 expression and activity have been observed in multiple neurological diseases. Here, we review recent findings to delineate the role of Rac1 signaling in neurodevelopmental disorders associated with abnormal spine morphology, synaptogenesis, and synaptic plasticity. Moreover, certain novel inhibitors of Rac1 and related pathways are discussed as potential avenues toward future treatment for these diseases.
Collapse
|
15
|
Parnell E, Shapiro LP, Voorn RA, Forrest MP, Jalloul HA, Loizzo DD, Penzes P. KALRN: A central regulator of synaptic function and synaptopathies. Gene 2020; 768:145306. [PMID: 33189799 DOI: 10.1016/j.gene.2020.145306] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/28/2020] [Accepted: 11/06/2020] [Indexed: 12/23/2022]
Abstract
The synaptic regulator, kalirin, plays a key role in synaptic plasticity and formation of dendritic arbors and spines. Dysregulation of the KALRN gene has been linked to various neurological disorders, including autism spectrum disorder, Alzheimer's disease, schizophrenia, addiction and intellectual disabilities. Both genetic and molecular studies highlight the importance of normal KALRN expression for healthy neurodevelopment and function. This review aims to give an in-depth analysis of the structure and molecular mechanisms of kalirin function, particularly within the brain. These data are correlated to genetic evidence of patient mutations within KALRN and animal models of Kalrn that together give insight into the manner in which this gene may be involved in neurodevelopment and the etiology of disease. The emerging links to human disease from post-mortem, genome wide association (GWAS) and exome sequencing studies are examined to highlight the disease relevance of kalirin, particularly in neurodevelopmental diseases. Finally, we will discuss efforts to pharmacologically regulate kalirin protein activity and the implications of such endeavors for the treatment of human disease. As multiple disease states arise from deregulated synapse formation and altered KALRN expression and function, therapeutics may be developed to provide control over KALRN activity and thus synapse dysregulation. As such, a detailed understanding of how kalirin regulates neuronal development, and the manner in which kalirin dysfunction promotes neurological disease, may support KALRN as a valuable therapeutic avenue for future pharmacological intervention.
Collapse
Affiliation(s)
- Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA
| | - Lauren P Shapiro
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA
| | - Roos A Voorn
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA
| | - Marc P Forrest
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA
| | - Hiba A Jalloul
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA
| | - Daniel D Loizzo
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, 60611 IL, USA; Northwestern University Center for Autism and Neurodevelopment, Chicago, IL 60611, USA.
| |
Collapse
|
16
|
Synaptic Kalirin-7 and Trio Interactomes Reveal a GEF Protein-Dependent Neuroligin-1 Mechanism of Action. Cell Rep 2020; 29:2944-2952.e5. [PMID: 31801062 PMCID: PMC9012321 DOI: 10.1016/j.celrep.2019.10.115] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/18/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
The RhoGEFs Kalirin-7 and Trio are regulators of synaptic plasticity, and their dysregulation is associated with a range of neurodevelopmental and neurodegenerative disorders. Although studies have implicated both Kalirin and Trio in certain diseases, such as tauopathies, they remarkably differ in their association with other disorders. Using unbiased proteomics, we identified interactomes of Kalirin-7 and Trio to ascertain distinct protein association networks associated with their respective function and revealed groups of proteins that preferentially interact with a particular RhoGEF. In comparison, we find Trio interacts with a range of axon guidance and presynaptic complexes, whereas Kalirin-7 associates with several synaptic adhesion molecules. Specifically, we show Kalirin-7 is an interactor of the cell adhesion molecule neuroligin-1 (NLGN1), and NLGN1-dependent synaptic function is mediated through Kalirin-7 in an interaction-dependent manner. Our data reveal not only the interactomes of two important disease-related proteins, but also provide an intracellular effector of NLGN1 function. Paskus et al. use quantitative proteomics to determine the synaptic interactomes of the disease-associated proteins Kalirin-7 and Trio, identifying Kalirin-7 as an interactor of NLGN1. Investigation of this interaction unveils Kalirin-7 as a primary intracellular effector of NLGN1 gain of function.
Collapse
|
17
|
Lu JH, Zhang MB, Wang JW, Ye XY, Chen JL, Jia GL, Xie CS, Shen YJ, Tao YX, Li J, Cao H. Kalirin-7 contributes to type 2 diabetic neuropathic pain via the postsynaptic density-95/N-methyl-D-aspartate receptor 2B-dependent N-methyl-D-aspartate receptor 2B phosphorylation in the spinal cord in rats. Am J Transl Res 2020; 12:4819-4829. [PMID: 32913553 PMCID: PMC7476141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/18/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE Diabetic neuropathic pain (DNP) is one of the common complications in type 2 Diabetes Mellitus (DM) patients. However, molecular mechanisms in underlying diabetic neuropathic pain are still poorly understood. Kalirin-7, a multifunctional Rho GDP/GTP exchange factor, located at the excitatory synapses, was reported to modulate the neuronal cytoskeleton. Therefore, in this study, we explored the effects of Kalirin-7 on type 2 diabetic neuropathic pain and the mechanisms in spinal cord in rats. METHODS The type 2 diabetic neuropathic pain model was established in rats by feeding them with a high-sugar and high-fat diet for 8 weeks, and then fasting them for 12 hours, followed by a single intraperitoneal injection of STZ. Kalirin-7 was knocked down in the spinal cord by an intrathecal administration of Kalirin-7 siRNA. RESULTS The levels of Kalirin-7, p-NR2B and PSD-95 as well as the PSD-95-NR2B coupling were significantly increased in the spinal cord of type 2 DM rats. The knockdown of Kalirin-7 expression in the spinal cord by the intrathecal administration of Kalirin-7 siRNA not only reduced the levels of p-NR2B and the PSD-95-NR2B coupling in the spinal cord, but also relieved mechanical allodynia and thermal hyperalgesia in type 2 DM rats. CONCLUSIONS Our findings suggest that spinally expressed Kalirin-7 likely contributes to type 2 diabetic neuropathic pain through regulating the PSD-95/NR2B interaction-dependent NR2B phosphorylation in the spinal cord.
Collapse
Affiliation(s)
- Jia-Hui Lu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Mao-Biao Zhang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Jun-Wu Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Xiu-Ying Ye
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Jia-Li Chen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Gai-Li Jia
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Ci-Shan Xie
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Yu-Jing Shen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New JerseyNewark, New Jersey 07103, USA
| | - Jun Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| | - Hong Cao
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical UniversityWenzhou 325035, Zhejiang, China
| |
Collapse
|
18
|
Kou XL, Tao Y, Xian JY, Lin YH, Cai CY, Wu HY, Chang L, Zhu DY. Uncoupling nNOS-PSD-95 in mPFC inhibits morphine priming-induced reinstatement after extinction training. Biochem Biophys Res Commun 2020; 525:520-527. [PMID: 32113678 DOI: 10.1016/j.bbrc.2020.02.112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 12/27/2022]
Abstract
Extremely high relapse rate is the dramatic challenge of drug abuse at present. Environmental cues play an important role in relapse of drug abuse. However, the specific mechanism underlying relapse remains unclear. Using morphine conditioned place preference (CPP) model, we show that association of neuronal nitric oxide synthase (nNOS) with postsynaptic density-95 (PSD-95) plays a significant role in morphine priming-induced reinstatement. The nNOS-PSD-95 coupling and c-Fos expression in the medial prefrontal cortex (mPFC) was significantly increased after extinction of morphine CPP. Dissociation of nNOS-PSD-95 in the mPFC by ZL006 inhibited the reinstatement of morphine CPP induced by a priming dose of morphine. Significantly reduced phosphorylation of cAMP-response element binding protein (CREB) in the mPFC was observed in the mice exposed to morphine after the extinction training. Uncoupling nNOS-PSD-95 reversed the morphine-induced CREB dysfunction. Moreover, effects of ZL006 on the reinstatement of morphine CPP and CREB activation depended on nNOS-PSD-95 target. Together, our findings suggest that nNOS-PSD-95 in the mPFC contributes to reinstatement of morphine CPP, possibly through CREB dysfunction, offering a potential target to prevent relapse of drug abuse.
Collapse
Affiliation(s)
- Xiao-Lin Kou
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Yan Tao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Jia-Yun Xian
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Cheng-Yun Cai
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
19
|
Li MX, Qiao H, Zhang M, Ma XM. Role of Cdk5 in Kalirin7-Mediated Formation of Dendritic Spines. Neurochem Res 2019; 44:1243-1251. [PMID: 30875016 DOI: 10.1007/s11064-019-02771-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 02/06/2023]
Abstract
A majority of excitatory synapses in the brain are localized on the dendritic spines. Alterations of spine density and morphology are associated with many neurological diseases. Understanding the molecular mechanisms underlying spine formation is important for understanding these diseases. Kalirin7 (Kal-7) is localized to the postsynaptic side of excitatory synapses in the neurons. Overexpression of Kal-7 causes an increase in spine density whereas knockdown expression of endogenous Kal-7 results in a decrease in spine density in primary cultured cortical neurons. However, the mechanisms underlying Kal-7-mediated spine formation are not entirely clear. Cyclin-dependent kinase 5 (Cdk5) plays a vital role in the formation of spines and synaptic plasticity. Kal-7 is phosphorylated by CDK5 at Thr1590, the unique Cdk5 phosphorylation site in the Kal-7 protein. This study was to explore the role of CDK5-mediated phosphorylation of Kal-7 in spine formation and the underlying mechanisms. Our results showed expression of Kal-7T/D (mimicked phosphorylation), Kal-7T/A mutants (blocked phosphorylation) or wild-type (Wt) Kal-7 caused in a similar increase in spine density, while spine size of Wt Kal-7-expressing cortical neurons was bigger than that in Kal-7 T\A-expressing neurons, but smaller than that in Kal-7T/D-expressing neurons. The fluorescence intensity of NMDA receptor subunit NR2B (GluN2B) staining was stronger along the MAP2 positive dendrites of Kal-7T/D-expressing neurons than that in Kal-7T/A- or Wt Kal-7-expressing neurons. The fluorescence intensity of AMPA receptor subunit GluR1 (GluA1) staining showed the same trend as GluN2B staining. These findings suggest that Cdk5 affects the function of Kal-7 on spine morphology and function via GluN2B and GluA1 receptors during dendritic spine formation.
Collapse
Affiliation(s)
- Ming-Xing Li
- State Key Laboratory of Subtropical Agro-Bioresource Conservation and Utilization, Guangxi University, Nanning, 530004, Guangxi, China
- College of Life Science, Shaanxi Normal University, Xi'an, 710062, Shaanxi, China
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Hui Qiao
- College of Life Science, Shaanxi Normal University, Xi'an, 710062, Shaanxi, China
| | - Ming Zhang
- State Key Laboratory of Subtropical Agro-Bioresource Conservation and Utilization, Guangxi University, Nanning, 530004, Guangxi, China
| | - Xin-Ming Ma
- College of Life Science, Shaanxi Normal University, Xi'an, 710062, Shaanxi, China.
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
20
|
Xie Z, Shapiro LP, Cahill ME, Russell TA, Lacor PN, Klein WL, Penzes P. Kalirin-7 prevents dendritic spine dysgenesis induced by amyloid beta-derived oligomers. Eur J Neurosci 2019; 49:1091-1101. [PMID: 30565792 DOI: 10.1111/ejn.14311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/19/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022]
Abstract
Synapse degeneration and dendritic spine dysgenesis are believed to be crucial early steps in Alzheimer's disease (AD), and correlate with cognitive deficits in AD patients. Soluble amyloid beta (Aβ)-derived oligomers, also termed Aβ-derived diffusible ligands (ADDLs), accumulate in the brain of AD patients and play a crucial role in AD pathogenesis. ADDLs bind to mature hippocampal neurons, induce structural changes in dendritic spines and contribute to neuronal death. However, mechanisms underlying structural and toxic effects are not fully understood. Here, we report that ADDLs bind to cultured mature cortical pyramidal neurons and induce spine dysgenesis. ADDL treatment induced the rapid depletion of kalirin-7, a brain-specific guanine-nucleotide exchange factor for the small GTPase Rac1, from spines. Kalirin-7 is a key regulator of dendritic spine morphogenesis and maintenance in forebrain pyramidal neurons and here we show that overexpression of kalirin-7 prevents ADDL-induced spine degeneration. Taken together, our results suggest that kalirin-7 may play a role in the early events leading to synapse degeneration, and its pharmacological activation may prevent or delay synapse pathology in AD.
Collapse
Affiliation(s)
- Zhong Xie
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Lauren P Shapiro
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Michael E Cahill
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Theron A Russell
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Pascale N Lacor
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois
| | - William L Klein
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois
| | - Peter Penzes
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
21
|
Motodate R, Saito H, Sobu Y, Hata S, Saito Y, Nakaya T, Suzuki T. X11 and X11-like proteins regulate the level of extrasynaptic glutamate receptors. J Neurochem 2018; 148:480-498. [PMID: 30411795 DOI: 10.1111/jnc.14623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Abstract
X11/Mint 1 and X11-like (X11L)/Mint 2 are neuronal adaptor protein to regulate trafficking and/or localization of various membrane proteins. By analyzing the localization of neuronal membrane proteins in X11-, X11L-, and X11/X11L doubly deficient mice with membrane fractionation procedures, we found that deficient of X11 and X11L decreased the level of glutamate receptors in non-PSD fraction. This finding suggests that X11 and X11L regulate the glutamate receptor micro-localization to the extrasynaptic region. In vitro coimmunoprecipitation studies of NMDA receptors lacking various cytoplasmic regions with X11 and X11L proteins harboring domain deletion suggest that extrasynaptic localization of NMDA receptor may be as a result of the multiple interactions of the receptor subunits with X11 and X11L regulated by protein phosphorylation, while that of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunits is not dependent on the binding with X11 and X11L proteins. Because the loss of X11 and X11L tends to impair the exocytosis, but not endocytosis, of glutamate receptors, NMDA receptors are likely to be supplied to the extrasynaptic plasma membrane with a way distinct from the mechanism regulating the localization of NMDA receptors into synaptic membrane region. Reduced localization of NMDA receptor into the extrasynaptic region increased slightly the phosphorylation level of cAMP responsible element binding protein in brain of X11/X11L doubly deficient mice compare to wild-type mice, suggesting a possible role of X11 and X11L in the regulation of signal transduction pathway through extrasynaptic glutamate receptors. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Rika Motodate
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Haruka Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Yuhki Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Tadashi Nakaya
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| |
Collapse
|
22
|
Modulation of Kalirin-7 expression by hippocampal CA1 5-HT 1B receptors in spatial memory consolidation. Behav Brain Res 2018; 356:148-155. [PMID: 29949735 DOI: 10.1016/j.bbr.2018.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 12/18/2022]
Abstract
Serotonin 5-HT1B receptors (5-HT1BRs) are distributed in hippocampal CA1 and play a pivotal role in cognitive function. Activation of 5-HT1BRs regulates synaptic plasticity at the excitatory synapses in the hippocampus. However, the role and its underlying mechanism of 5-HT1BR activation-mediated glutamatergic synaptic plasticity in spatial memory are not fully understood. In this study, spatial memory of Sprague-Dawley (SD) rats was assessed in a Morris water maze after bilateral dorsal hippocampal CA1 infusion of the 5-HT1BR antagonist GR55562 (25 μg/μL) or agonist CP93129 (25 μg/μL). GR55562 did not affect the spatial memory acquisition but significantly increased the target quadrant preference during the memory consolidation probe performed 14 d after the training session, while CP93129 impaired the memory consolidation process. Moreover, GR55562 significantly increased, while CP93129 significantly decreased, the density of dendritic spines on the distal apical dendrites of CA1 pyramidal neurons. Furthermore, western blot experiments indicated that GR55562 significantly increased, but CP93129 significantly reduced, the expression of Kalirin-7 (Kal-7), PSD95, and GluA2/3 subunits of AMPA receptors. Our results suggest that Kal-7 and Kal-7-mediatedalteration of AMPA receptor subtype expression may play crucial roles in the impact of hippocampal CA1 5-HT1BR activation on spatial memory consolidation.
Collapse
|
23
|
Cissé M, Duplan E, Lorivel T, Dunys J, Bauer C, Meckler X, Gerakis Y, Lauritzen I, Checler F. The transcription factor XBP1s restores hippocampal synaptic plasticity and memory by control of the Kalirin-7 pathway in Alzheimer model. Mol Psychiatry 2017; 22:1562-1575. [PMID: 27646263 PMCID: PMC5658671 DOI: 10.1038/mp.2016.152] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/14/2016] [Accepted: 07/18/2016] [Indexed: 12/18/2022]
Abstract
Neuronal network dysfunction and cognitive decline constitute the most prominent features of Alzheimer's disease (AD), although mechanisms causing such impairments are yet to be determined. Here we report that virus-mediated delivery of the active spliced transcription factor X-Box binding protein 1s (XBP1s) in the hippocampus rescued spine density, synaptic plasticity and memory function in a mouse model of AD. XBP1s transcriptionally activated Kalirin-7 (Kal7), a protein that controls synaptic plasticity. In addition, we found reduced levels of Kal7 in primary neurons exposed to Aβ oligomers, transgenic mouse models and human AD brains. Short hairpin RNA-mediated knockdown of Kal7 altered synaptic plasticity and memory formation in naive mice. Further, reduction of endogenous Kal7 compromised the beneficial effects of XBP1s in Alzheimer's model. Hence, our findings reveal that XBP1s is neuroprotective through a mechanism that engages Kal7 pathway with therapeutic implications in AD pathology.
Collapse
Affiliation(s)
- M Cissé
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France,Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, NEUROLOGY, IPMC/CNRS, 660 Route des Lucioles, 06560 Valbonne, France. E-mail:
| | - E Duplan
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - T Lorivel
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - J Dunys
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - C Bauer
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - X Meckler
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - Y Gerakis
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - I Lauritzen
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - F Checler
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
24
|
Hui S, Yang Y, Peng WJ, Sheng CX, Gong W, Chen S, Xu PP, Wang Z. Protective effects of Bushen Tiansui decoction on hippocampal synapses in a rat model of Alzheimer's disease. Neural Regen Res 2017; 12:1680-1686. [PMID: 29171433 PMCID: PMC5696849 DOI: 10.4103/1673-5374.217347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2017] [Indexed: 12/26/2022] Open
Abstract
Bushen Tiansui decoction is composed of six traditional Chinese medicines: Herba Epimedii, Radix Polygoni multiflori, Plastrum testudinis, Fossilia Ossis Mastodi, Radix Polygalae, and Rhizoma Acorus tatarinowii. Because Bushen Tiansui decoction is effective against amyloid beta (Aβ) toxicity, we hypothesized that it would reduce hippocampal synaptic damage and improve cognitive function in Alzheimer's disease. To test this hypothesis, we used a previously established animal model of Alzheimer's disease, that is, microinjection of aggregated Aβ25-35 into the bilateral brain ventricles of Sprague-Dawley rats. We found that long-term (28 days) oral administration of Bushen Tiansui decoction (0.563, 1.688, and 3.375 g/mL; 4 mL/day) prevented synaptic loss in the hippocampus and increased the expression levels of synaptic proteins, including postsynaptic density protein 95, the N-methyl-D-aspartate receptor 2B subunit, and Shank1. These results suggested that Bushen Tiansui decoction can protect synapses by maintaining the expression of these synaptic proteins. Bushen Tiansui decoction also ameliorated measures reflecting spatial learning and memory deficits that were observed in the Morris water maze (i.e., increased the number of platform crossings and the amount of time spent in the target quadrant and decreased escape latency) following intraventricular injections of aggregated Aβ25-35 compared with those measures in untreated Aβ25-35-injected rats. Overall, these results provided evidence that further studies on the prevention and treatment of dementia with this traditional Chinese medicine are warranted.
Collapse
Affiliation(s)
- Shan Hui
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Geriatric Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu Yang
- Department of Geriatric Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei-jun Peng
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chen-xia Sheng
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei Gong
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Shuai Chen
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Pan-pan Xu
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhe Wang
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
25
|
Miller MB, Yan Y, Machida K, Kiraly DD, Levy AD, Wu YI, Lam TT, Abbott T, Koleske AJ, Eipper BA, Mains RE. Brain Region and Isoform-Specific Phosphorylation Alters Kalirin SH2 Domain Interaction Sites and Calpain Sensitivity. ACS Chem Neurosci 2017; 8:1554-1569. [PMID: 28418645 DOI: 10.1021/acschemneuro.7b00076] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Kalirin7 (Kal7), a postsynaptic Rho GDP/GTP exchange factor (RhoGEF), plays a crucial role in long-term potentiation and in the effects of cocaine on behavior and spine morphology. The KALRN gene has been linked to schizophrenia and other disorders of synaptic function. Mass spectrometry was used to quantify phosphorylation at 26 sites in Kal7 from individual adult rat nucleus accumbens and prefrontal cortex before and after exposure to acute or chronic cocaine. Region- and isoform-specific phosphorylation was observed along with region-specific effects of cocaine on Kal7 phosphorylation. Evaluation of the functional significance of multisite phosphorylation in a complex protein like Kalirin is difficult. With the identification of five tyrosine phosphorylation (pY) sites, a panel of 71 SH2 domains was screened, identifying subsets that interacted with multiple pY sites in Kal7. In addition to this type of reversible interaction, endoproteolytic cleavage by calpain plays an essential role in long-term potentiation. Calpain cleaved Kal7 at two sites, separating the N-terminal domain, which affects spine length, and the PDZ binding motif from the GEF domain. Mutations preventing phosphorylation did not affect calpain sensitivity or GEF activity; phosphomimetic mutations at specific sites altered protein stability, increased calpain sensitivity, and reduced GEF activity.
Collapse
Affiliation(s)
| | | | | | - Drew D. Kiraly
- Department
of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | | | | | | | | | | | | | | |
Collapse
|
26
|
LaRese TP, Yan Y, Eipper BA, Mains RE. Using Kalirin conditional knockout mice to distinguish its role in dopamine receptor mediated behaviors. BMC Neurosci 2017; 18:45. [PMID: 28535798 PMCID: PMC5442696 DOI: 10.1186/s12868-017-0363-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/19/2017] [Indexed: 01/25/2023] Open
Abstract
Background Mice lacking Kalirin-7 (Kal7KO), a Rho GDP/GTP exchange factor, self-administer cocaine at a higher rate than wildtype mice, and show an exaggerated locomotor response to experimenter-administered cocaine. Kal7, which localizes to post-synaptic densities at glutamatergic synapses, interacts directly with the GluN2B subunit of the N-methyl-d-aspartate (NMDA; GluN) receptor. Consistent with these observations, Kal7 plays an essential role in NMDA receptor dependent long term potentiation and depression, and glutamatergic transmission plays a key role in the response to chronic cocaine. A number of genetic studies have implicated altered Kalirin expression in schizophrenia and other disorders such as Alzheimer’s Disease. Results A comparison of the effects of experimenter-administered cocaine on mice lacking all Kalirin isoforms to its effects on mice lacking only Kalirin-7 identified Kal7 as the key isoform whose deletion produces exaggerated locomotor responses to cocaine. Pretreatment of Kal7KO mice with a low dose of ifenprodil, a selective GluN2B antagonist, eliminated their enhanced locomotor response to cocaine, revealing an important role for GluN2B in this behavior. Selective knockout of Kalirin in dopamine transporter expressing neurons produced a transient enhancement of cocaine-induced locomotion, while knockout of Kalirin in Drd1a- or Drd2-dopamine receptor expressing neurons was without effect. As observed in Kalirin global knockout mice, eliminating Kalirin expression in Drd2-expressing neurons increased exploratory behavior in the elevated zero maze, an effect eliminated by pretreatment with ifenprodil. Conclusions The cocaine-sensitive neuronal pathways which are most sensitive to altered Kalirin function may be the pathways most dependent on GluN2B and Drd2. Electronic supplementary material The online version of this article (doi:10.1186/s12868-017-0363-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taylor P LaRese
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030-3401, USA
| | - Yan Yan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030-3401, USA
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030-3401, USA.,Departments of Neuroscience and Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030-3401, USA
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030-3401, USA.
| |
Collapse
|
27
|
Qiao H, An SC, Xu C, Ma XM. Role of proBDNF and BDNF in dendritic spine plasticity and depressive-like behaviors induced by an animal model of depression. Brain Res 2017; 1663:29-37. [PMID: 28284898 DOI: 10.1016/j.brainres.2017.02.020] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
|
28
|
Cissé M, Duplan E, Checler F. The transcription factor XBP1 in memory and cognition: Implications in Alzheimer disease. Mol Med 2017; 22:905-917. [PMID: 28079229 DOI: 10.2119/molmed.2016.00229] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022] Open
Abstract
X-box binding protein 1 (XBP1) is a unique basic region leucine zipper transcription factor isolated two decades ago in a search for regulators of major histocompatibility complex class II gene expression. XBP1 is a very complex protein regulating many physiological functions, including immune system, inflammatory responses, and lipid metabolism. Evidence over the past few years suggests that XBP1 also plays important roles in pathological settings since its activity as transcription factor has profound effects on the prognosis and progression of diseases such as cancer, neurodegeneration, and diabetes. Here we provide an overview on recent advances in our understanding of this multifaceted molecule, particularly in regulating synaptic plasticity and memory function, and the implications in neurodegenerative diseases with emphasis on Alzheimer disease.
Collapse
Affiliation(s)
- Moustapha Cissé
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Eric Duplan
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
29
|
Abstract
For more than 20 years, we have known that Ca(2+)/calmodulin-dependent protein kinase (CaMKII) activation is both necessary and sufficient for the induction of long-term potentiation (LTP). During this time, tremendous effort has been spent in attempting to understand how CaMKII activation gives rise to this phenomenon. Despite such efforts, there is much to be learned about the molecular mechanisms involved in LTP induction downstream of CaMKII activation. In this review, we highlight recent developments that have shaped our current thinking about the molecular mechanisms underlying LTP and discuss important questions that remain in the field.
Collapse
Affiliation(s)
| | - Roger A Nicoll
- Department of Cellular and Molecular Pharmacology and.,Department of Physiology, University of California, San Francisco, California 94143; ,
| |
Collapse
|
30
|
Miller MB, Yan Y, Wu Y, Hao B, Mains RE, Eipper BA. Alternate promoter usage generates two subpopulations of the neuronal RhoGEF Kalirin-7. J Neurochem 2016; 140:889-902. [PMID: 27465683 DOI: 10.1111/jnc.13749] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 11/28/2022]
Abstract
Kalirin (Kal), a dual Rho GDP/GTP exchange factor (GEF), plays essential roles within and outside the nervous system. Tissue-specific, developmentally regulated alternative splicing generates isoforms with one (Kal7) or two (Kal9, Kal12) GEF domains along with a kinase (Kal12) domain; while Kal9 and Kal12 are crucial for neurite outgrowth, Kal7 plays important roles in spine maintenance and synaptic plasticity. Tissue-specific usage of alternate Kalrn promoters (A, B, C, D) places four different peptides before the Sec14 domain. cSec14, with an amphipathic helix encoded by the C-promoter (Kal-C-helix), is the only variant known to interact with phosphoinositides. We sought to elucidate the biological significance of Kalirin promoter usage and lipid binding. While Ex1B expression was predominant early in development, Ex1C expression increased when synaptogenesis occurred. Kal-C-helix-containing Kal7 (cKal7) was enriched at the postsynaptic density, present in the microsomal fraction and absent from cytosol; no significant amount of cKal9 or cKal12 could be identified in mouse brain. Similarly, in primary hippocampal neurons, endogenous cKalirin colocalized with postsynaptic density 95 in dendritic spines, juxtaposed to Vglut1-positive puncta. When expressed in young neurons, bSec14-EGFP was diffusely distributed, while cSec14-EGFP localized to internal puncta. Transfected bKal7-EGFP and cKal7-EGFP localized to dendritic spines and increased spine density in more mature cultured neurons. Although promoter usage did not alter the Rac-GEF activity of Kal7, the synaptic puncta formed by cKal7-EGFP were smaller than those formed by bKal7-EGFP. Molecular modeling predicted a role for Kal-C-helix residue Arg15 in the interaction of cSec14 with phosphoinositides. Consistent with this prediction, mutation of Arg15 to Gln altered the localization of cSec14-EGFP and cKal7-EGFP. These data suggest that phosphoinositide-dependent interactions unique to cKal7 contribute to protein localization and function. Cover Image for this issue: doi. 10.1111/jnc.13791.
Collapse
Affiliation(s)
- Megan B Miller
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Yan Yan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Yi Wu
- Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Bing Hao
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, USA.,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
31
|
Kalirin is required for BDNF-TrkB stimulated neurite outgrowth and branching. Neuropharmacology 2016; 107:227-238. [PMID: 27036892 DOI: 10.1016/j.neuropharm.2016.03.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/03/2016] [Accepted: 03/28/2016] [Indexed: 01/19/2023]
Abstract
Exogenous brain-derived neurotrophic factor (BDNF), acting through TrkB, is known to promote neurite formation and branching. This response to BDNF was eliminated by inhibition of TrkB kinase and by specific inhibition of the GEF1 domain of Kalirin, which activates Rac1. Neurons from Kalrn knockout mice were unable to activate Rac1 in response to BDNF. BDNF-triggered neurite outgrowth was abolished when Kalrn expression was reduced using shRNA that targets all of the major Kalrn isoforms, and reduced in neurons from Kalrn knockout mice. The Kalrn isoforms expressed early in development also include a GEF2 domain that activates RhoA. However, BDNF-stimulated neurite outgrowth in Kalrn knockout neurons was rescued by expression of Kalirin-7, which includes only the GEF1 domain but lacks the GEF2 domain. Dendritic morphogenesis, which requires spatially restricted, coordinated changes in the actin cytoskeleton and in the organization of microtubules, involves essential contributions from multiple Rho GEFs. Since Tiam1, another Rho GEF, is also required for BDNF-stimulated neurite outgrowth, an inhibitory fragment of Tiam1 (PHn-CC-EX) was tested and found to interfere with both Kalirin and Tiam1 GEF activity. The prolonged TrkB activation observed in response to BDNF in Kalrn knockout neurons and the altered time course and extent of ERK, CREB and Akt activation observed in the absence of Kalrn would be expected to alter the response of these neurons to other regulatory factors.
Collapse
|
32
|
Liddie S, Itzhak Y. Variations in the stimulus salience of cocaine reward influences drug-associated contextual memory. Addict Biol 2016; 21:242-54. [PMID: 25351485 DOI: 10.1111/adb.12191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drugs of abuse act as reinforcers because they influence learning and memory processes resulting in long-term memory of drug reward. We have previously shown that mice conditioned by fixed daily dose of cocaine (Fix-C) or daily escalating doses of cocaine (Esc-C) resulted in short- and long-term persistence of drug memory, respectively, suggesting different mechanisms in acquisition of cocaine memory. The present study was undertaken to investigate the differential contribution of N-methyl-D-aspartate receptor (NMDAR) subunits in the formation of Fix-C and Esc-C memory in C57BL/6J mice. Training by Esc-C resulted in marked elevation in hippocampal expression of Grin2b mRNA and NR2B protein levels compared with training by Fix-C. The NR2B-containing NMDAR antagonist ifenprodil had similar attenuating effects on acquisition and reconsolidation of Fix-C and Esc-C memory. However, the NMDAR antagonist MK-801 had differential effects: (1) higher doses of MK-801 were required for post-retrieval disruption of reconsolidation of Esc-C memory than Fix-C memory; and (2) pre-retrieval MK-801 inhibited extinction of Fix-C memory but it had no effect on Esc-C memory. In addition, blockade of NMDAR downstream signaling pathways also showed differential regulation of Fix-C and Esc-C memory. Inhibition of neuronal nitric oxide synthase attenuated acquisition and disrupted reconsolidation of Fix-C but not Esc-C memory. In contrast, the mitogen-activating extracellular kinase inhibitor SL327 attenuated reconsolidation of Esc-C but not Fix-C memory. These results suggest that NMDAR downstream signaling molecules associated with consolidation and reconsolidation of cocaine-associated memory may vary upon changes in the salience of cocaine reward during conditioning.
Collapse
Affiliation(s)
- Shervin Liddie
- Division of Neuroscience; University of Miami Miller School of Medicine; Miami FL USA
| | - Yossef Itzhak
- Department of Psychiatry and Behavioral Sciences; University of Miami Miller School of Medicine; Miami FL USA
- Division of Neuroscience; University of Miami Miller School of Medicine; Miami FL USA
| |
Collapse
|
33
|
Elagabani MN, Briševac D, Kintscher M, Pohle J, Köhr G, Schmitz D, Kornau HC. Subunit-selective N-Methyl-d-aspartate (NMDA) Receptor Signaling through Brefeldin A-resistant Arf Guanine Nucleotide Exchange Factors BRAG1 and BRAG2 during Synapse Maturation. J Biol Chem 2016; 291:9105-18. [PMID: 26884337 DOI: 10.1074/jbc.m115.691717] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Indexed: 11/06/2022] Open
Abstract
The maturation of glutamatergic synapses in the CNS is regulated by NMDA receptors (NMDARs) that gradually change from a GluN2B- to a GluN2A-dominated subunit composition during postnatal development. Here we show that NMDARs control the activity of the small GTPase ADP-ribosylation factor 6 (Arf6) by consecutively recruiting two related brefeldin A-resistant Arf guanine nucleotide exchange factors, BRAG1 and BRAG2, in a GluN2 subunit-dependent manner. In young cortical cultures, GluN2B and BRAG1 tonically activated Arf6. In mature cultures, Arf6 was activated through GluN2A and BRAG2 upon NMDA treatment, whereas the tonic Arf6 activation was not detectable any longer. This shift in Arf6 regulation and the associated drop in Arf6 activity were reversed by a knockdown of BRAG2. Given their sequential recruitment during development, we examined whether BRAG1 and BRAG2 influence synaptic currents in hippocampal CA1 pyramidal neurons using patch clamp recordings in acute slices from mice at different ages. The number of AMPA receptor (AMPAR) miniature events was reduced by depletion of BRAG1 but not by depletion of BRAG2 during the first 2 weeks after birth. In contrast, depletion of BRAG2 during postnatal weeks 4 and 5 reduced the number of AMPAR miniature events and compromised the quantal sizes of both AMPAR and NMDAR currents evoked at Schaffer collateral synapses. We conclude that both Arf6 activation through GluN2B-BRAG1 during early development and the transition from BRAG1- to BRAG2-dependent Arf6 signaling induced by the GluN2 subunit switch are critical for the development of mature glutamatergic synapses.
Collapse
Affiliation(s)
- Mohammad Nael Elagabani
- From the Neuroscience Research Center (NWFZ) and Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| | - Dušica Briševac
- From the Neuroscience Research Center (NWFZ) and Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| | | | - Jörg Pohle
- the Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Georg Köhr
- the Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | | | - Hans-Christian Kornau
- From the Neuroscience Research Center (NWFZ) and Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| |
Collapse
|
34
|
Ba W, Yan Y, Reijnders MRF, Schuurs-Hoeijmakers JHM, Feenstra I, Bongers EMHF, Bosch DGM, De Leeuw N, Pfundt R, Gilissen C, De Vries PF, Veltman JA, Hoischen A, Mefford HC, Eichler EE, Vissers LELM, Nadif Kasri N, De Vries BBA. TRIO loss of function is associated with mild intellectual disability and affects dendritic branching and synapse function. Hum Mol Genet 2015; 25:892-902. [PMID: 26721934 DOI: 10.1093/hmg/ddv618] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/18/2015] [Indexed: 12/21/2022] Open
Abstract
Recently, we marked TRIO for the first time as a candidate gene for intellectual disability (ID). Across diverse vertebrate species, TRIO is a well-conserved Rho GTPase regulator that is highly expressed in the developing brain. However, little is known about the specific events regulated by TRIO during brain development and its clinical impact in humans when mutated. Routine clinical diagnostic testing identified an intragenic de novo deletion of TRIO in a boy with ID. Targeted sequencing of this gene in over 2300 individuals with ID, identified three additional truncating mutations. All index cases had mild to borderline ID combined with behavioral problems consisting of autistic, hyperactive and/or aggressive behavior. Studies in dissociated rat hippocampal neurons demonstrated the enhancement of dendritic formation by suppressing endogenous TRIO, and similarly decreasing endogenous TRIO in organotypic hippocampal brain slices significantly increased synaptic strength by increasing functional synapses. Together, our findings provide new mechanistic insight into how genetic deficits in TRIO can lead to early neuronal network formation by directly affecting both neurite outgrowth and synapse development.
Collapse
Affiliation(s)
- Wei Ba
- Department of Human Genetics, Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Yan Yan
- Department of Neuroscience, UCONN Health Center, Farmington, CT 06030, USA
| | | | | | | | | | - Daniëlle G M Bosch
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Bartiméus, Institute for the Visually Impaired, Zeist, The Netherlands and
| | | | | | | | | | - Joris A Veltman
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | - Heather C Mefford
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA, Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | | | - Nael Nadif Kasri
- Department of Human Genetics, Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands,
| | - Bert B A De Vries
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands,
| |
Collapse
|
35
|
Carlson K, Pomerantz SC, Vafa O, Naso M, Strohl W, Mains RE, Eipper BA. Optimizing production of Fc-amidated peptides by Chinese hamster ovary cells. BMC Biotechnol 2015; 15:95. [PMID: 26475607 PMCID: PMC4609047 DOI: 10.1186/s12896-015-0210-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 10/01/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amidation of the carboxyl terminal of many peptides is essential for full biological potency, often increasing receptor binding and stability. The single enzyme responsible for this reaction is peptidylglycine α-amidating monooxygenase (PAM: EC 1.14.17.3), a copper- and ascorbate-dependent Type I membrane protein. METHODS To make large amounts of high molecular weight amidated product, Chinese hamster ovary (CHO) cells were engineered to express exogenous PAM. To vary access of the enzyme to its substrate, exogenous PAM was targeted to the endoplasmic reticulum, trans-Golgi network, endosomes and lysosomes or to the lumen of the secretory pathway. RESULTS PAM was equally active when targeted to each intracellular location and assayed in homogenates. Immunocytochemical analyses of CHO cells and a pituitary cell line demonstrated that targeting of exogenous PAM was partially successful. PAM substrates generated by expressing peptidylglycine substrates (glucagon-like peptide 1-Gly, peptide YY-Gly and neuromedin U-Gly) fused to the C-terminus of immunoglobulin Fc in CHO cell lines producing targeted PAM. The extent of amidation of the Fc-peptides was determined by mass spectrometry and amidation-specific enzyme immunoassays. Amidation was inhibited by copper chelation, but was not enhanced by the addition of additional copper or ascorbate. CONCLUSIONS Peptide amidation was increased over endogenous levels by exogenous PAM, and targeting PAM to the endoplasmic reticulum or trans-Golgi network increased peptide amidation compared to endogenous CHO PAM.
Collapse
Affiliation(s)
- Kristina Carlson
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| | - Steven C Pomerantz
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - Omid Vafa
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - Michael Naso
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - William Strohl
- Biologics Research, Biotechnology Center of Excellence, Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA. .,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
36
|
A role for Kalirin-7 in nociceptive sensitization via activity-dependent modulation of spinal synapses. Nat Commun 2015; 6:6820. [PMID: 25865668 PMCID: PMC4403379 DOI: 10.1038/ncomms7820] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/03/2015] [Indexed: 12/20/2022] Open
Abstract
Synaptic plasticity is the cornerstone of processes underlying persistent nociceptive activity-induced changes in normal nociceptive sensitivity. Kalirin-7 is a multifunctional guanine-nucleotide-exchange factor (GEF) for Rho GTPases that is characterized by its localization at excitatory synapses, interactions with glutamate receptors and its ability to dynamically modulate the neuronal cytoskeleton. Here we show that spinally expressed Kalirin-7 is required for persistent nociceptive activity-dependent synaptic long-term potentiation as well as activity-dependent remodelling of synaptic spines in the spinal dorsal horn, thereby orchestrating functional and structural plasticity during the course of inflammatory pain.
Collapse
|
37
|
Miller MB, Vishwanatha KS, Mains RE, Eipper BA. An N-terminal Amphipathic Helix Binds Phosphoinositides and Enhances Kalirin Sec14 Domain-mediated Membrane Interactions. J Biol Chem 2015; 290:13541-55. [PMID: 25861993 DOI: 10.1074/jbc.m115.636746] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Indexed: 11/06/2022] Open
Abstract
Previous studies revealed an essential role for the lipid-binding Sec14 domain of kalirin (KalSec14), but its mechanism of action is not well understood. Because alternative promoter usage appends unique N-terminal peptides to the KalSec14 domain, we used biophysical, biochemical, and cell biological approaches to examine the two major products, bKalSec14 and cKalSec14. Promoter B encodes a charged, unstructured peptide, whereas promoter C encodes an amphipathic helix (Kal-C-helix). Both bKalSec14 and cKalSec14 interacted with lipids in PIP strip and liposome flotation assays, with significantly greater binding by cKalSec14 in both assays. Disruption of the hydrophobic face of the Kal-C-helix in cKalSec14KKED eliminated its increased liposome binding. Although cKalSec14 showed significantly reduced binding to liposomes lacking phosphatidylinositol phosphates or cholesterol, liposome binding by bKalSec14 and cKalSec14KKED was not affected. When expressed in AtT-20 cells, bKalSec14-GFP was diffusely localized, whereas cKalSec14-GFP localized to the trans-Golgi network and secretory granules. The amphipathic C-helix was sufficient for this localization. When AtT-20 cells were treated with a cell-permeant derivative of the Kal-C-helix (Kal-C-helix-Arg9), we observed increased secretion of a product stored in mature secretory granules, with no effect on basal secretion; a cell-permeant control peptide (Kal-C-helixKKED-Arg9) did not have this effect. Through its ability to control expression of a novel, phosphoinositide-binding amphipathic helix, Kalrn promoter usage is expected to affect function.
Collapse
Affiliation(s)
| | | | | | - Betty A Eipper
- From the Departments of Neuroscience and Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut 06030
| |
Collapse
|
38
|
Regulating Rac in the nervous system: molecular function and disease implication of Rac GEFs and GAPs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:632450. [PMID: 25879033 PMCID: PMC4388020 DOI: 10.1155/2015/632450] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/06/2015] [Indexed: 12/11/2022]
Abstract
Rho family GTPases, including RhoA, Rac1, and Cdc42 as the most studied members, are master regulators of actin cytoskeletal organization. Rho GTPases control various aspects of the nervous system and are associated with a number of neuropsychiatric and neurodegenerative diseases. The activity of Rho GTPases is controlled by two families of regulators, guanine nucleotide exchange factors (GEFs) as the activators and GTPase-activating proteins (GAPs) as the inhibitors. Through coordinated regulation by GEFs and GAPs, Rho GTPases act as converging signaling molecules that convey different upstream signals in the nervous system. So far, more than 70 members of either GEFs or GAPs of Rho GTPases have been identified in mammals, but only a small subset of them have well-known functions. Thus, characterization of important GEFs and GAPs in the nervous system is crucial for the understanding of spatiotemporal dynamics of Rho GTPase activity in different neuronal functions. In this review, we summarize the current understanding of GEFs and GAPs for Rac1, with emphasis on the molecular function and disease implication of these regulators in the nervous system.
Collapse
|
39
|
Qiao H, An SC, Ren W, Ma XM. Progressive alterations of hippocampal CA3-CA1 synapses in an animal model of depression. Behav Brain Res 2014; 275:191-200. [PMID: 25192638 DOI: 10.1016/j.bbr.2014.08.040] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 08/08/2014] [Accepted: 08/20/2014] [Indexed: 12/11/2022]
Abstract
Major depressive disorder is the most prevalent psychiatric condition, but the cellular and molecular mechanisms underlying this disorder are largely unknown, although multiple hypotheses have been proposed. The aim of this study was to characterize the progressive alteration of neuronal plasticity in the male rat hippocampus during depression induced by chronic unpredictable mild stress (CUMS), an established animal model of depression. The data in the hippocampus were collected on days 7, 14 and 21 after the onset of three-week CUMS. When analyzed on day 21, three-week CUMS induced typically depressive-like behaviors, impaired LTP induction, and decreased basal synaptic transmission at hippocampal CA3-CA1 synapses recorded in vivo, which was accompanied by decreased density of dendritic spines in CA1 and CA3 pyramidal neurons. The levels of both Kalirin-7 and brain-derived neurotrophic factor (BDNF) in the hippocampus were decreased at the same time. On day 14 (middle phase), some depressive-like behaviors were observed, which was accompanied by depressed basal synaptic transmission and enhanced LTP induction at the CA3-CA1 synapses. However, BDNF expression was decreased without alteration of Kalirin7 expression in comparison with no-stress control. Depressed basal synaptic transmission occurred in the middle phase of CUMS may contribute to decreased expression of BDNF. On day 7, depressive-like behaviors were not observed, and LTP induction, spine density, Kalirin-7 and BDNF expression were not altered by CUMS in comparison with no-stress control. These results showed that the functional changes at CA3-CA1synapses occurred earlier than the structural alteration during three-week CUMS as a strategy of neural adaptation, and rats required three weeks to develop depressive-like behaviors during CUMS. Our results suggest an important role of Kalirin-7 in CUMS-mediated alterations in spine density, synaptic function and overall depressive-like behaviors on day 21.
Collapse
Affiliation(s)
- Hui Qiao
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China
| | - Shu-Cheng An
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China.
| | - Wei Ren
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China
| | - Xin-Ming Ma
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China; University of Connecticut Health Center, Department of Neuroscience, Farmington, CT 06030, USA
| |
Collapse
|
40
|
Cissé M, Checler F. Eph receptors: new players in Alzheimer's disease pathogenesis. Neurobiol Dis 2014; 73:137-49. [PMID: 25193466 DOI: 10.1016/j.nbd.2014.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is devastating and leads to permanent losses of memory and other cognitive functions. Although recent genetic evidences strongly argue for a causative role of Aβ in AD onset and progression (Jonsson et al., 2012), its role in AD etiology remains a matter of debate. However, even if not the sole culprit or pathological trigger, genetic and anatomical evidences in conjunction with numerous pharmacological studies, suggest that Aβ peptides, at least contribute to the disease. How Aβ contributes to memory loss remains largely unknown. Soluble Aβ species referred to as Aβ oligomers have been shown to be neurotoxic and induce network failure and cognitive deficits in animal models of the disease. In recent years, several proteins were described as potential Aβ oligomers receptors, amongst which are the receptor tyrosine kinases of Eph family. These receptors together with their natural ligands referred to as ephrins have been involved in a plethora of physiological and pathological processes, including embryonic neurogenesis, learning and memory, diabetes, cancers and anxiety. Here we review recent discoveries on Eph receptors-mediated protection against Aβ oligomers neurotoxicity as well as their potential as therapeutic targets in AD pathogenesis.
Collapse
Affiliation(s)
- Moustapha Cissé
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| |
Collapse
|
41
|
Li J, Zhang S, Zhang L, Wang R, Wang M. Effects of l-3-n-Butylphthalide on Cognitive Dysfunction and NR2B Expression in Hippocampus of Streptozotocin (STZ)-Induced Diabetic Rats. Cell Biochem Biophys 2014; 71:315-22. [DOI: 10.1007/s12013-014-0200-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
42
|
Mandela P, Yan Y, LaRese T, Eipper BA, Mains RE. Elimination of Kalrn expression in POMC cells reduces anxiety-like behavior and contextual fear learning. Horm Behav 2014; 66:430-8. [PMID: 25014196 PMCID: PMC4127147 DOI: 10.1016/j.yhbeh.2014.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 11/17/2022]
Abstract
Kalirin, a Rho GDP/GTP exchange factor for Rac1 and RhoG, is known to play an essential role in the formation and maintenance of excitatory synapses and in the secretion of neuropeptides. Mice unable to express any of the isoforms of Kalrn in cells that produce POMC at any time during development (POMC cells) exhibited reduced anxiety-like behavior and reduced acquisition of passive avoidance behavior, along with sex-specific alteration in the corticosterone response to restraint stress. Strikingly, lack of Kalrn expression in POMC cells closely mimicked the effects of global Kalrn knockout on anxiety-like behavior and passive avoidance conditioning without causing the other deficits noted in Kalrn knockout mice. Our data suggest that deficits in excitatory inputs onto POMC neurons are responsible for the behavioral phenotypes observed.
Collapse
Affiliation(s)
- Prashant Mandela
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030-3401, United States
| | - Yan Yan
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030-3401, United States
| | - Taylor LaRese
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030-3401, United States
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030-3401, United States
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030-3401, United States.
| |
Collapse
|
43
|
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
|
44
|
Kolodziejczyk K, Parsons MP, Southwell AL, Hayden MR, Raymond LA. Striatal synaptic dysfunction and hippocampal plasticity deficits in the Hu97/18 mouse model of Huntington disease. PLoS One 2014; 9:e94562. [PMID: 24728353 PMCID: PMC3984157 DOI: 10.1371/journal.pone.0094562] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/18/2014] [Indexed: 01/01/2023] Open
Abstract
Huntington disease (HD) is a fatal neurodegenerative disorder caused by a CAG repeat expansion in the gene (HTT) encoding the huntingtin protein (HTT). This mutation leads to multiple cellular and synaptic alterations that are mimicked in many current HD animal models. However, the most commonly used, well-characterized HD models do not accurately reproduce the genetics of human disease. Recently, a new ‘humanized’ mouse model, termed Hu97/18, has been developed that genetically recapitulates human HD, including two human HTT alleles, no mouse Hdh alleles and heterozygosity of the HD mutation. Previously, behavioral and neuropathological testing in Hu97/18 mice revealed many features of HD, yet no electrophysiological measures were employed to investigate possible synaptic alterations. Here, we describe electrophysiological changes in the striatum and hippocampus of the Hu97/18 mice. At 9 months of age, a stage when cognitive deficits are fully developed and motor dysfunction is also evident, Hu97/18 striatal spiny projection neurons (SPNs) exhibited small changes in membrane properties and lower amplitude and frequency of spontaneous excitatory postsynaptic currents (sEPSCs); however, release probability from presynaptic terminals was unaltered. Strikingly, these mice also exhibited a profound deficiency in long-term potentiation (LTP) at CA3-to-CA1 synapses. In contrast, at 6 months of age we found only subtle alterations in SPN synaptic transmission, while 3-month old animals did not display any electrophysiologically detectable changes in the striatum and CA1 LTP was intact. Together, these data reveal robust, progressive deficits in synaptic function and plasticity in Hu97/18 mice, consistent with previously reported behavioral abnormalities, and suggest an optimal age (9 months) for future electrophysiological assessment in preclinical studies of HD.
Collapse
Affiliation(s)
- Karolina Kolodziejczyk
- Department of Psychiatry, Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew P. Parsons
- Department of Psychiatry, Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amber L. Southwell
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael R. Hayden
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lynn A. Raymond
- Department of Psychiatry, Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
45
|
Hippocampal long-term potentiation is disrupted during expression and extinction but is restored after reinstatement of morphine place preference. J Neurosci 2014; 34:527-38. [PMID: 24403152 DOI: 10.1523/jneurosci.2838-13.2014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Learned associations between environmental cues and morphine use play an important role in the maintenance and/or relapse of opioid addiction. Although previous studies suggest that context-dependent morphine treatment alters glutamatergic transmission and synaptic plasticity in the hippocampus, their role in morphine conditioned place preference (CPP) and reinstatement remains unknown. We investigated changes in synaptic plasticity and NMDAR expression in the hippocampus after the expression, extinction, and reinstatement of morphine CPP. Here we report that morphine CPP is associated with increased basal synaptic transmission, impaired hippocampal long-term potentiation (LTP), and increased synaptic expression of the NR1 and NR2b NMDAR subunits. Changes in synaptic plasticity, synaptic NR1 and NR2b expression, and morphine CPP were absent when morphine was not paired with a specific context. Furthermore, hippocampal LTP was impaired and synaptic NR2b expression was increased after extinction of morphine CPP, indicating that these alterations in plasticity may be involved in the mechanisms underlying the learning of drug-environment associations. After extinction of morphine CPP, a priming dose of morphine was sufficient to reinstate morphine CPP and was associated with LTP that was indistinguishable from saline control groups. In contrast, morphine CPP extinguished mice that received a saline priming dose did not show CPP and had disrupted hippocampal LTP. Finally, we found that reinstatement of morphine CPP was prevented by the selective blockade of the NR2b subunit in the hippocampus. Together, these data suggest that alterations in synaptic plasticity and glutamatergic transmission play an important role in the reinstatement of morphine CPP.
Collapse
|
46
|
Kiraly DD, Nemirovsky NE, LaRese TP, Tomek SE, Yahn SL, Olive MF, Eipper BA, Mains RE. Constitutive knockout of kalirin-7 leads to increased rates of cocaine self-administration. Mol Pharmacol 2013; 84:582-90. [PMID: 23894151 PMCID: PMC3781382 DOI: 10.1124/mol.113.087106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/26/2013] [Indexed: 01/01/2023] Open
Abstract
Kalirin-7 (Kal7) is a Rho-guanine nucleotide exchange factor that is localized in neuronal postsynaptic densities. Kal7 interacts with the NR2B subunit of the NMDA receptor and regulates aspects of dendritic spine dynamics both in vitro and in vivo. Chronic treatment with cocaine increases dendritic spine density in the nucleus accumbens (NAc) of rodents and primates. Kal7 mRNA and protein are upregulated in the NAc following cocaine treatment, and the presence of Kal7 is necessary for the normal proliferation of dendritic spines following cocaine use. Mice that constitutively lack Kal7 [Kalirin-7 knockout mice (Kal7(KO))] demonstrate increased locomotor sensitization to cocaine and a decreased place preference for cocaine. Here, using an intravenous cocaine self-administration paradigm, Kal7(KO) mice exhibit increased administration of cocaine at lower doses as compared with wild-type (Wt) mice. Analyses of mRNA transcript levels from the NAc of mice that self-administered saline or cocaine reveal that larger splice variants of the Kalrn gene are increased by cocaine more dramatically in Kal7(KO) mice than in Wt mice. Additionally, transcripts encoding the NR2B subunit of the NMDA receptor increased in Wt mice that self-administered cocaine but were unchanged in similarly experienced Kal7(KO) mice. These findings suggest that Kal7 participates in the reinforcing effects of cocaine, and that Kal7 and cocaine interact to alter the expression of genes related to critical glutamatergic signaling pathways in the NAc.
Collapse
Affiliation(s)
- Drew D Kiraly
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut (D.D.K., T.P.L., B.A.E., R.E.M.); and Department of Psychology, Program in Behavioral Neuroscience (N.E.N., S.E.T., S.L.Y., M.F.O.) and Interdisciplinary Graduate Program in Neuroscience (M.F.O.), Arizona State University, Tempe, Arizona
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kalirin-7 mediates cocaine-induced AMPA receptor and spine plasticity, enabling incentive sensitization. J Neurosci 2013; 33:11012-22. [PMID: 23825406 DOI: 10.1523/jneurosci.1097-13.2013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is well established that behavioral sensitization to cocaine is accompanied by increased spine density and AMPA receptor (AMPAR) transmission in the nucleus accumbens (NAc), but two major questions remain unanswered. Are these adaptations mechanistically coupled? And, given that they can be dissociated from locomotor sensitization, what is their functional significance? We tested the hypothesis that the guanine-nucleotide exchange factor Kalirin-7 (Kal-7) couples cocaine-induced AMPAR and spine upregulation and that these adaptations underlie sensitization of cocaine's incentive-motivational properties-the properties that make it "wanted." Rats received eight daily injections of saline or cocaine. On withdrawal day 14, we found that Kal-7 levels and activation of its downstream effectors Rac-1 and PAK were increased in the NAc of cocaine-sensitized rats. Furthermore, AMPAR surface expression and spine density were increased, as expected. To determine whether these changes require Kal-7, a lentiviral vector expressing Kal-7 shRNA was injected into the NAc core before cocaine exposure. Knocking down Kal-7 abolished the AMPAR and spine upregulation normally seen during cocaine withdrawal. Despite the absence of these adaptations, rats with reduced Kal-7 levels developed locomotor sensitization. However, incentive sensitization, which was assessed by how rapidly rats learned to self-administer a threshold dose of cocaine, was severely impaired. These results identify a signaling pathway coordinating AMPAR and spine upregulation during cocaine withdrawal, demonstrate that locomotor and incentive sensitization involve divergent mechanisms, and link enhanced excitatory transmission in the NAc to incentive sensitization.
Collapse
|
48
|
Spinal serum-inducible and glucocorticoid-inducible kinase 1 mediates neuropathic pain via kalirin and downstream PSD-95-dependent NR2B phosphorylation in rats. J Neurosci 2013; 33:5227-40. [PMID: 23516288 DOI: 10.1523/jneurosci.4452-12.2013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The coupling of the spinal postsynaptic density-95 (PSD-95) with the glutamatergic N-methyl-d-aspartate receptor NR2B subunit and the subsequent NR2B phosphorylation contribute to pain-related plasticity. Increasing evidence reveals that kalirin, a Rho-guanine nucleotide exchange factor, modulates PSD-95-NR2B-dependent neuroplasticity. Our laboratory recently demonstrated that serum-inducible and glucocorticoid-inducible kinase 1 (SGK1) participates in inflammation-associated pain hypersensitivity by modulating spinal glutamatergic neurotransmission. Because kalirin is one of the proteins in PSD that is highly phosphorylated by various kinases, we tested whether kalirin could be a downstream target of spinal SGK1 that participates in neuropathic pain development via regulation of the PSD-95-NR2B coupling-dependent phosphorylation of NR2B. We observed that spinal nerve ligation (SNL, L5) in male Sprague Dawley rats resulted in behavioral allodynia, which was associated with phosphorylated SGK1 (pSGK1), kalirin, and phosphorylated NR2B (pNR2B) expression and an increase in pSGK1-kalirin-PSD-95-pNR2B coprecipitation in the ipsilateral dorsal horn (L4-L5). SNL-enhanced kalirin immunofluorescence was coincident with pSGK1, PSD-95, and pNR2B immunoreactivity. Small-interfering RNA (siRNA) that targeted spinal kalirin mRNA expression (10 μg, 10 μl; i.t.) reduced SNL-induced allodynia, kalirin and pNR2B expression, as well as kalirin-PSD-95 and PSD-95-pNR2B coupling and costaining without affecting SGK1 phosphorylation. Daily administration of GSK-650394 (an SGK1 antagonist; 100 nm, 10 μl, i.t.) not only exhibited effects similar to the kalirin mRNA-targeting siRNA but also attenuated pSGK1-kalirin costaining and SGK1-kalirin coupling. We suggest that nerve injury could induce spinal SGK1 phosphorylation that subsequently interacts with and upregulates kalirin to participate in neuropathic pain development via PSD-95-NR2B coupling-dependent NR2B phosphorylation.
Collapse
|
49
|
Gao C, Tronson NC, Radulovic J. Modulation of behavior by scaffolding proteins of the post-synaptic density. Neurobiol Learn Mem 2013; 105:3-12. [PMID: 23701866 DOI: 10.1016/j.nlm.2013.04.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/05/2013] [Accepted: 04/09/2013] [Indexed: 12/23/2022]
Abstract
Scaffolding proteins of the neuronal post-synaptic density (PSD) are principal organizers of glutamatergic neurotransmission that bring together glutamate receptors and signaling molecules at discrete synaptic locations. Genetic alterations of individual PSD scaffolds therefore disrupt the function of entire multiprotein modules rather than a single glutamatergic mechanism, and thus induce a range of molecular and structural abnormalities in affected neurons. Despite such broad molecular consequences, knockout, knockdown, or knockin of glutamate receptor scaffolds typically affect a subset of specific behaviors and thereby mold and specialize the actions of the ubiquitous glutamatergic neurotransmitter system. Approaches designed to control the function of neuronal scaffolds may therefore have high potential to restore behavioral morbidities and comorbidities in patients with psychiatric disorders. Here we summarize a series of experiments with genetically modified mice revealing the roles of main N-methyl-d-aspartate (NMDA) and group I metabotropic glutamate (mGluR1/5) receptor scaffolds in behavior, discuss the clinical implications of the findings, and propose future research directions.
Collapse
Affiliation(s)
- Can Gao
- Jiangsu Key Laboratory of Anesthesiology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | | | | |
Collapse
|
50
|
Postsynaptic density scaffold SAP102 regulates cortical synapse development through EphB and PAK signaling pathway. J Neurosci 2013; 33:5040-52. [PMID: 23486974 DOI: 10.1523/jneurosci.2896-12.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Membrane-associated guanylate kinases (MAGUKs), including SAP102, PSD-95, PSD-93, and SAP97, are scaffolding proteins for ionotropic glutamate receptors at excitatory synapses. MAGUKs play critical roles in synaptic plasticity; however, details of signaling roles for each MAGUK remain largely unknown. Here we report that SAP102 regulates cortical synapse development through the EphB and PAK signaling pathways. Using lentivirus-delivered shRNAs, we found that SAP102 and PSD-95, but not PSD-93, are necessary for excitatory synapse formation and synaptic AMPA receptor (AMPAR) localization in developing mouse cortical neurons. SAP102 knockdown (KD) increased numbers of elongated dendritic filopodia, which is often observed in mouse models and human patients with mental retardation. Further analysis revealed that SAP102 coimmunoprecipitated the receptor tyrosine kinase EphB2 and RacGEF Kalirin-7 in neonatal cortex, and SAP102 KD reduced surface expression and dendritic localization of EphB. Moreover, SAP102 KD prevented reorganization of actin filaments, synapse formation, and synaptic AMPAR trafficking in response to EphB activation triggered by its ligand ephrinB. Last, p21-activated kinases (PAKs) were downregulated in SAP102 KD neurons. These results demonstrate that SAP102 has unique roles in cortical synapse development by mediating EphB and its downstream PAK signaling pathway. Both SAP102 and PAKs are associated with X-linked mental retardation in humans; thus, synapse formation mediated by EphB/SAP102/PAK signaling in the early postnatal brain may be crucial for cognitive development.
Collapse
|