1
|
Mezzacappa C, Komiya Y, Habas R. Reversion induced LIM domain protein (RIL) is a Daam1-interacting protein and regulator of the actin cytoskeleton during non-canonical Wnt signaling. Dev Biol 2024; 515:46-58. [PMID: 38968989 PMCID: PMC11321505 DOI: 10.1016/j.ydbio.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024]
Abstract
The Daam1 protein regulates Wnt-induced cytoskeletal changes during vertebrate gastrulation though its full mode of action and binding partners remain unresolved. Here we identify Reversion Induced LIM domain protein (RIL) as a new interacting protein of Daam1. Interaction studies uncover binding of RIL to the C-terminal actin-nucleating portion of Daam1 in a Wnt-responsive manner. Immunofluorescence studies showed subcellular localization of RIL to actin fibers and co-localization with Daam1 at the plasma membrane. RIL gain- and loss-of-function approaches in Xenopus produced severe gastrulation defects in injected embryos. Additionally, a simultaneous loss of Daam1 and RIL synergized to produce severe gastrulation defects indicating RIL and Daam1 may function in the same signaling pathway. RIL further synergizes with another novel Daam1-interacting protein, Formin Binding Protein 1 (FNBP1), to regulate gastrulation. Our studies altogether show RIL mediates Daam1-regulated non-canonical Wnt signaling that is required for vertebrate gastrulation.
Collapse
Affiliation(s)
| | - Yuko Komiya
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA
| | - Raymond Habas
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA.
| |
Collapse
|
2
|
Srivastava Y, Donta M, Mireles LL, Paulucci-Holthauzen A, Shi L, Bedford MT, Waxham MN, McCrea PD. Exploring the PDZ, DUF, and LIM Domains of Pdlim5 in Dendrite Branching. Int J Mol Sci 2024; 25:8326. [PMID: 39125895 PMCID: PMC11312917 DOI: 10.3390/ijms25158326] [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: 05/18/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The branched architecture of neuronal dendrites is a key factor in how neurons form ordered networks and discoveries continue to be made identifying proteins and protein-protein interactions that direct or execute the branching and extension of dendrites. Our prior work showed that the molecular scaffold Pdlim5 and delta-catenin, in conjunction, are two proteins that help regulate the branching and elongation of dendrites in cultured hippocampal neurons and do so through a phosphorylation-dependent mechanism triggered by upstream glutamate signaling. In this report we have focused on Pdlim5's multiple scaffolding domains and how each contributes to dendrite branching. The three identified regions within Pdlim5 are the PDZ, DUF, and a trio of LIM domains; however, unresolved is the intra-molecular conformation of Pdlim5 as well as which domains are essential to regulate dendritic branching. We address Pdlim5's structure and function by examining the role of each of the domains individually and using deletion mutants in the context of the full-length protein. Results using primary hippocampal neurons reveal that the Pdlim5 DUF domain plays a dominant role in increasing dendritic branching. Neither the PDZ domain nor the LIM domains alone support increased branching. The central role of the DUF domain was confirmed using deletion mutants in the context of full-length Pdlim5. Guided by molecular modeling, additional domain mapping studies showed that the C-terminal LIM domain forms a stable interaction with the N-terminal PDZ domain, and we identified key amino acid residues at the interface of each domain that are needed for this interaction. We posit that the central DUF domain of Pdlim5 may be subject to modulation in the context of the full-length protein by the intra-molecular interaction between the N-terminal PDZ and C-terminal LIM domains. Overall, our studies reveal a novel mechanism for the regulation of Pdlim5's function in the regulation of neuronal branching and highlight the critical role of the DUF domain in mediating these effects.
Collapse
Affiliation(s)
- Yogesh Srivastava
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maxsam Donta
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics & Epigenetics, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| | - Lydia L. Mireles
- Department of Neurobiology & Anatomy, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| | | | - Leilei Shi
- Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mark T. Bedford
- Program in Genetics & Epigenetics, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
- Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - M. Neal Waxham
- Department of Neurobiology & Anatomy, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
- Program in Neuroscience, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| | - Pierre D. McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics & Epigenetics, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
- Program in Neuroscience, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| |
Collapse
|
3
|
Cao YY, Wu LL, Li XN, Yuan YL, Zhao WW, Qi JX, Zhao XY, Ward N, Wang J. Molecular Mechanisms of AMPA Receptor Trafficking in the Nervous System. Int J Mol Sci 2023; 25:111. [PMID: 38203282 PMCID: PMC10779435 DOI: 10.3390/ijms25010111] [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: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Synaptic plasticity enhances or reduces connections between neurons, affecting learning and memory. Postsynaptic AMPARs mediate greater than 90% of the rapid excitatory synaptic transmission in glutamatergic neurons. The number and subunit composition of AMPARs are fundamental to synaptic plasticity and the formation of entire neural networks. Accordingly, the insertion and functionalization of AMPARs at the postsynaptic membrane have become a core issue related to neural circuit formation and information processing in the central nervous system. In this review, we summarize current knowledge regarding the related mechanisms of AMPAR expression and trafficking. The proteins related to AMPAR trafficking are discussed in detail, including vesicle-related proteins, cytoskeletal proteins, synaptic proteins, and protein kinases. Furthermore, significant emphasis was placed on the pivotal role of the actin cytoskeleton, which spans throughout the entire transport process in AMPAR transport, indicating that the actin cytoskeleton may serve as a fundamental basis for AMPAR trafficking. Additionally, we summarize the proteases involved in AMPAR post-translational modifications. Moreover, we provide an overview of AMPAR transport and localization to the postsynaptic membrane. Understanding the assembly, trafficking, and dynamic synaptic expression mechanisms of AMPAR may provide valuable insights into the cognitive decline associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yang Cao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Ling-Ling Wu
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Xiao-Nan Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Yu-Lian Yuan
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Wan-Wei Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Jing-Xuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Xu-Yu Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Natalie Ward
- Medical Laboratory, Exceptional Community Hospital, 19060 N John Wayne Pkwy, Maricopa, AZ 85139, USA;
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| |
Collapse
|
4
|
Healy MD, Collins BM. The PDLIM family of actin-associated proteins and their emerging role in membrane trafficking. Biochem Soc Trans 2023; 51:2005-2016. [PMID: 38095060 PMCID: PMC10754285 DOI: 10.1042/bst20220804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
The PDZ and LIM domain (PDLIM) proteins are associated with the actin cytoskeleton and have conserved in roles in metazoan actin organisation and function. They primarily function as scaffolds linking various proteins to actin and its binding partner α-actinin via two conserved domains; an N-terminal postsynaptic density 95, discs large and zonula occludens-1 (PDZ) domain, and either single or multiple C-terminal LIN-11, Isl-1 and MEC-3 (LIM) domains in the actinin-associated LIM protein (ALP)- and Enigma-related proteins, respectively. While their role in actin organisation, such as in stress fibres or in the Z-disc of muscle fibres is well known, emerging evidence also suggests a role in actin-dependent membrane trafficking in the endosomal system. This is mediated by a recently identified interaction with the sorting nexin 17 (SNX17) protein, an adaptor for the trafficking complex Commander which is itself intimately linked to actin-directed formation of endosomal recycling domains. In this review we focus on the currently understood structural basis for PDLIM function. The PDZ domains mediate direct binding to distinct classes of PDZ-binding motifs (PDZbms), including α-actinin and other actin-associated proteins, and a highly specific interaction with the type III PDZbm such as the one found in the C-terminus of SNX17. The structures of the LIM domains are less well characterised and how they engage with their ligands is completely unknown. Despite the lack of experimental structural data, we find that recently developed machine learning-based structure prediction methods provide insights into their potential interactions and provide a template for further studies of their molecular functions.
Collapse
Affiliation(s)
- Michael D. Healy
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| | - Brett M. Collins
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| |
Collapse
|
5
|
Wang Y, Wang J, Yan Z, Liu S, Xu W. Potential drug targets for asthma identified in the plasma and brain through Mendelian randomization analysis. Front Immunol 2023; 14:1240517. [PMID: 37809092 PMCID: PMC10551444 DOI: 10.3389/fimmu.2023.1240517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Background Asthma is a heterogeneous disease, and the involvement of neurogenic inflammation is crucial in its development. The standardized treatments focus on alleviating symptoms. Despite the availability of medications for asthma, they have proven to be inadequate in controlling relapses and halting the progression of the disease. Therefore, there is a need for novel drug targets to prevent asthma. Methods We utilized Mendelian randomization to investigate potential drug targets for asthma. We analyzed summary statistics from the UK Biobank and then replicated our findings in GWAS data by Demenais et al. and the FinnGen cohort. We obtained genetic instruments for 734 plasma and 73 brain proteins from recently reported GWAS. Next, we utilized reverse causal relationship analysis, Bayesian co-localization, and phenotype scanning as part of our sensitivity analysis. Furthermore, we performed a comparison and protein-protein interaction analysis to identify causal proteins. We also analyzed the possible consequences of our discoveries by the given existing asthma drugs and their targets. Results Using Mendelian randomization analysis, we identified five protein-asthma pairs that were significant at the Bonferroni level (P < 6.35 × 10-5). Specifically, in plasma, we found that an increase of one standard deviation in IL1R1 and ECM1 was associated with an increased risk of asthma, while an increase in ADAM19 was found to be protective. The corresponding odds ratios were 1.03 (95% CI, 1.02-1.04), 1.00 (95% CI, 1.00-1.01), and 0.99 (95% CI, 0.98-0.99), respectively. In the brain, per 10-fold increase in ECM1 (OR, 1.05; 95% CI, 1.03-1.08) and PDLIM4 (OR, 1.05; 95% CI, 1.04-1.07) increased the risk of asthma. Bayesian co-localization found that ECM1 in the plasma (coloc.abf-PPH4 = 0.965) and in the brain (coloc.abf-PPH4 = 0.931) shared the same mutation with asthma. The target proteins of current asthma medications were found to interact with IL1R1. IL1R1 and PDLIM4 were validated in two replication cohorts. Conclusion Our integrative analysis revealed that asthma risk is causally affected by the levels of IL1R1, ECM1, and PDLIM4. The results suggest that these three proteins have the potential to be used as drug targets for asthma, and further investigation through clinical trials is needed.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Otorhinolaryngology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jiaxi Wang
- Department of Otorhinolaryngology, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhanfeng Yan
- Department of Otorhinolaryngology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Siming Liu
- Department of Otorhinolaryngology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wenlong Xu
- Department of Otorhinolaryngology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Proteomic identification and structural basis for the interaction between sorting nexin SNX17 and PDLIM family proteins. Structure 2022; 30:1590-1602.e6. [DOI: 10.1016/j.str.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 06/29/2022] [Accepted: 09/30/2022] [Indexed: 12/03/2022]
|
7
|
Fisher LAB, Schöck F. The unexpected versatility of ALP/Enigma family proteins. Front Cell Dev Biol 2022; 10:963608. [PMID: 36531944 PMCID: PMC9751615 DOI: 10.3389/fcell.2022.963608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
One of the most intriguing features of multicellular animals is their ability to move. On a cellular level, this is accomplished by the rearrangement and reorganization of the cytoskeleton, a dynamic network of filamentous proteins which provides stability and structure in a stationary context, but also facilitates directed movement by contracting. The ALP/Enigma family proteins are a diverse group of docking proteins found in numerous cellular milieus and facilitate these processes among others. In vertebrates, they are characterized by having a PDZ domain in combination with one or three LIM domains. The family is comprised of CLP-36 (PDLIM1), Mystique (PDLIM2), ALP (PDLIM3), RIL (PDLIM4), ENH (PDLIM5), ZASP (PDLIM6), and Enigma (PDLIM7). In this review, we will outline the evolution and function of their protein domains which confers their versatility. Additionally, we highlight their role in different cellular environments, focusing specifically on recent advances in muscle research using Drosophila as a model organism. Finally, we show the relevance of this protein family to human myopathies and the development of muscle-related diseases.
Collapse
|
8
|
Dutta P, Bharti P, Kumar J, Maiti S. Role of actin cytoskeleton in the organization and function of ionotropic glutamate receptors. Curr Res Struct Biol 2021; 3:277-289. [PMID: 34766008 PMCID: PMC8569634 DOI: 10.1016/j.crstbi.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/04/2021] [Accepted: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
Neural networks with precise connection are compulsory for learning and memory. Various cellular events occur during the genesis of dendritic spines to their maturation, synapse formation, stabilization of the synapse, and proper signal transmission. The cortical actin cytoskeleton and its multiple regulatory proteins are crucial for the above cellular events. The different types of ionotropic glutamate receptors (iGluRs) present on the postsynaptic density (PSD) are also essential for learning and memory. Interaction of the iGluRs in association of their auxiliary proteins with actin cytoskeleton regulated by actin-binding proteins (ABPs) are required for precise long-term potentiation (LTP) and long-term depression (LTD). There has been a quest to understand the mechanistic detail of synapse function involving these receptors with dynamic actin cytoskeleton. A major, emerging area of investigation is the relationship between ABPs and iGluRs in synapse development. In this review we have summarized the current understanding of iGluRs functioning with respect to the actin cytoskeleton, scaffolding proteins, and their regulators. The AMPA, NMDA, Delta and Kainate receptors need the stable underlying actin cytoskeleton to anchor through synaptic proteins for precise synapse formation. The different types of ABPs present in neurons play a critical role in dynamizing/stabilizing the actin cytoskeleton needed for iGluRs function.
Collapse
Affiliation(s)
- Priyanka Dutta
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Pratibha Bharti
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Janesh Kumar
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Sankar Maiti
- Indian Institute of Science Education and Research, Kolkata, 741246, India
| |
Collapse
|
9
|
Traceable stimulus-dependent rapid molecular changes in dendritic spines in the brain. Sci Rep 2020; 10:15266. [PMID: 32943708 PMCID: PMC7499203 DOI: 10.1038/s41598-020-72248-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/26/2020] [Indexed: 01/21/2023] Open
Abstract
Dendritic spines function as microcompartments that can modify the efficiency of their associated synapses. Here, we analyzed stimulus-dependent molecular changes in spines. The F-actin capping protein CapZ accumulates in parts of dendritic spines within regions where long-term potentiation has been induced. We produced a transgenic mouse line, AiCE-Tg, in which CapZ tagged with enhanced green fluorescence protein (EGFP-CapZ) is expressed. Twenty minutes after unilateral visual or somatosensory stimulation in AiCE-Tg mice, relative EGFP-CapZ signal intensification was seen in a subset of dendritic spines selectively in stimulated-side cortices; this right-left difference was abolished by NMDA receptor blockade. Immunolabeling of α-actinin, a PSD-95 binding protein that can recruit AMPA receptors, showed that the α-actinin signals colocalized more frequently in spines with the brightest EGFP-CapZ signals (top 100) than in spines with more typical EGFP-CapZ signal strength (top 1,000). This stimulus-dependent in vivo redistribution of EGFP-CapZ represents a novel molecular event with plasticity-like characteristics, and bright EGFP-CapZ in AiCE-Tg mice make high-CapZ spines traceable in vivo and ex vivo. This mouse line has the potential to be used to reveal sequential molecular events, including synaptic tagging, and to relate multiple types of plasticity in these spines, extending knowledge related to memory mechanisms.
Collapse
|
10
|
Turlova E, Wong R, Xu B, Li F, Du L, Habbous S, Horgen FD, Fleig A, Feng ZP, Sun HS. TRPM7 Mediates Neuronal Cell Death Upstream of Calcium/Calmodulin-Dependent Protein Kinase II and Calcineurin Mechanism in Neonatal Hypoxic-Ischemic Brain Injury. Transl Stroke Res 2020; 12:164-184. [PMID: 32430797 DOI: 10.1007/s12975-020-00810-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 11/25/2022]
Abstract
Transient receptor potential melastatin 7 (TRPM7), a calcium-permeable, ubiquitously expressed ion channel, is critical for axonal development, and mediates hypoxic and ischemic neuronal cell death in vitro and in vivo. However, the downstream mechanisms underlying the TRPM7-mediated processes in physiology and pathophysiology remain unclear. In this study, we employed a mouse model of hypoxic-ischemic brain cell death which mimics the pathophysiology of hypoxic-ischemic encephalopathy (HIE). HIE is a major public health issue and an important cause of neonatal deaths worldwide; however, the available treatments for HIE remain limited. Its survivors face life-long neurological challenges including mental retardation, cerebral palsy, epilepsy and seizure disorders, motor impairments, and visual and auditory impairments. Through a proteomic analysis, we identified calcium/calmodulin-dependent protein kinase II (CaMKII) and phosphatase calcineurin as potential mediators of cell death downstream from TRPM7 activation. Further analysis revealed that TRPM7 mediates cell death through CaMKII, calmodulin, calcineurin, p38, and cofilin cascade. In vivo, we found a significant reduction of brain injury and improvement of short- and long-term functional outcomes after HI after administration of specific TRPM7 blocker waixenicin A. Our data demonstrate a molecular mechanism of TRPM7-mediated cell death and identifies TRPM7 as a promising therapeutic and drug development target for HIE.
Collapse
Affiliation(s)
- Ekaterina Turlova
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Raymond Wong
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Baofeng Xu
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Feiya Li
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Lida Du
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Steven Habbous
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI, 96744, USA
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96720, USA
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| | - Hong-Shuo Sun
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Pharmacology, University of Toronto, 1 King's College Circle, Toronto, M5S 1A8, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, University of Toronto, Toronto, Canada.
| |
Collapse
|
11
|
Dendritic Spines in Alzheimer's Disease: How the Actin Cytoskeleton Contributes to Synaptic Failure. Int J Mol Sci 2020; 21:ijms21030908. [PMID: 32019166 PMCID: PMC7036943 DOI: 10.3390/ijms21030908] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by Aβ-driven synaptic dysfunction in the early phases of pathogenesis. In the synaptic context, the actin cytoskeleton is a crucial element to maintain the dendritic spine architecture and to orchestrate the spine’s morphology remodeling driven by synaptic activity. Indeed, spine shape and synaptic strength are strictly correlated and precisely governed during plasticity phenomena in order to convert short-term alterations of synaptic strength into long-lasting changes that are embedded in stable structural modification. These functional and structural modifications are considered the biological basis of learning and memory processes. In this review we discussed the existing evidence regarding the role of the spine actin cytoskeleton in AD synaptic failure. We revised the physiological function of the actin cytoskeleton in the spine shaping and the contribution of actin dynamics in the endocytosis mechanism. The internalization process is implicated in different aspects of AD since it controls both glutamate receptor membrane levels and amyloid generation. The detailed understanding of the mechanisms controlling the actin cytoskeleton in a unique biological context as the dendritic spine could pave the way to the development of innovative synapse-tailored therapeutic interventions and to the identification of novel biomarkers to monitor synaptic loss in AD.
Collapse
|
12
|
Ríos H, Paganelli AR, Fosser NS. The role of PDLIM1, a PDZ-LIM domain protein, at the ribbon synapses in the chicken retina. J Comp Neurol 2020; 528:1820-1832. [PMID: 31930728 DOI: 10.1002/cne.24855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 01/29/2023]
Abstract
PDLIM's protein family is involved in the rearrangement of the actin cytoskeleton. In the present study, we describe the localization of PDLIM1 in chicken photoreceptors. This study provides evidence that this protein is present at the cone pedicles, as well as in other synapses of the chicken retina. Here, we demonstrate the expression pattern of PDLIM1 through immunofluorescence staining, immunoblots, subcellular fractionation, and immunoprecipitation experiments. Also, we consider the possibility that PDLIM1 may be involved in the synaptic vesicle endocytosis and/or the presynaptic trafficking of synaptic vesicles back to the nonready releasable pool. This endocytotic/exocytotic coupling requires a tight link between exocytic vesicle fusion at defined release sites and endocytic retrieval of synaptic vesicle membranes. In turn, photoreceptor ribbon synaptic structure depends on the cytoskeleton arrangement, both at the active zone-related with exocytosis-as well as at the endocytic zone-periactive zone. To our knowledge, the PDLIM1 protein has not been observed in the pre synapses of the retina. Thus, the present study describes the expression and subcellular localization of PDLIM1 for the first time, as well as its modulation by visual environment in the chicken retina.
Collapse
Affiliation(s)
- Hugo Ríos
- Universidad de Buenos Aires, Facultad de Medicina, I° U.A. Histología, Embriología, Biología Celular y Genética, Ciudad de Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Alejandra R Paganelli
- Universidad de Buenos Aires, Facultad de Medicina, I° U.A. Histología, Embriología, Biología Celular y Genética, Ciudad de Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Nicolás S Fosser
- Universidad de Buenos Aires, Facultad de Medicina, I° U.A. Histología, Embriología, Biología Celular y Genética, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
13
|
PDLIM4/RIL-mediated regulation of Src and malignant properties of breast cancer cells. Oncotarget 2020; 11:22-30. [PMID: 32002121 PMCID: PMC6967772 DOI: 10.18632/oncotarget.27410] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/16/2019] [Indexed: 11/25/2022] Open
Abstract
RIL/PDLIM4 gene was identified as a tumor suppressor, its expression is frequently altered in various types of malignancies. The product of RIL/PDLIM4 gene is an adapter protein involved in the actin cytoskeleton remolding and assembly of stress fibers crucial for cell motility and epithelial-mesenchymal transition. Although the exact mechanism tethering RIL to cancer development remains unknown some pieces of evidence suggest that RIL may act by suppressing activation of the proto-oncogene tyrosine-protein kinase Src. To further explore this issue we tested how different expression levels of RIL affected the activity of Src in breast cancer cell lines. RIL was ectopically overexpressed in the cell cultures with its relatively low endogenous level, or, otherwise, was downregulated by RNA interference. Whereas we observed no correlation between expression levels of RIL and activity of Src we found that in several cell lines elevated levels of RIL were associated with higher cell migratory activity along with the increased incidence of breast xenograft formation and metastasizing. The obtained data suggest that in some breast cancer models RIL may not act as Src kinase inhibitor, but rather play the role of a potential oncogene that promotes cell motility and contributes to cancer cells spreading.
Collapse
|
14
|
Tousley A, Iuliano M, Weisman E, Sapp E, Richardson H, Vodicka P, Alexander J, Aronin N, DiFiglia M, Kegel-Gleason KB. Huntingtin associates with the actin cytoskeleton and α-actinin isoforms to influence stimulus dependent morphology changes. PLoS One 2019; 14:e0212337. [PMID: 30768638 PMCID: PMC6377189 DOI: 10.1371/journal.pone.0212337] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
One response of cells to growth factor stimulus involves changes in morphology driven by the actin cytoskeleton and actin associated proteins which regulate functions such as cell adhesion, motility and in neurons, synaptic plasticity. Previous studies suggest that Huntingtin may be involved in regulating morphology however, there has been limited evidence linking endogenous Huntingtin localization or function with cytoplasmic actin in cells. We found that depletion of Huntingtin in human fibroblasts reduced adhesion and altered morphology and these phenotypes were made worse with growth factor stimulation, whereas the presence of the Huntington's Disease mutation inhibited growth factor induced changes in morphology and increased numbers of vinculin-positive focal adhesions. Huntingtin immunoreactivity localized to actin stress fibers, vinculin-positive adhesion contacts and membrane ruffles in fibroblasts. Interactome data from others has shown that Huntingtin can associate with α-actinin isoforms which bind actin filaments. Mapping studies using a cDNA encoding α-actinin-2 showed that it interacts within Huntingtin aa 399-969. Double-label immunofluorescence showed Huntingtin and α-actinin-1 co-localized to stress fibers, membrane ruffles and lamellar protrusions in fibroblasts. Proximity ligation assays confirmed a close molecular interaction between Huntingtin and α-actinin-1 in human fibroblasts and neurons. Huntingtin silencing with siRNA in fibroblasts blocked the recruitment of α-actinin-1 to membrane foci. These studies support the idea that Huntingtin is involved in regulating adhesion and actin dependent functions including those involving α-actinin.
Collapse
Affiliation(s)
- Adelaide Tousley
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Maria Iuliano
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Elizabeth Weisman
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Ellen Sapp
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Heather Richardson
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Petr Vodicka
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Jonathan Alexander
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Neil Aronin
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Marian DiFiglia
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Kimberly B. Kegel-Gleason
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
15
|
Tseng HY, Samarelli AV, Kammerer P, Scholze S, Ziegler T, Immler R, Zent R, Sperandio M, Sanders CR, Fässler R, Böttcher RT. LCP1 preferentially binds clasped αMβ2 integrin and attenuates leukocyte adhesion under flow. J Cell Sci 2018; 131:jcs.218214. [PMID: 30333137 DOI: 10.1242/jcs.218214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Integrins are α/β heterodimers that interconvert between inactive and active states. In the active state the α/β cytoplasmic domains recruit integrin-activating proteins and separate the transmembrane and cytoplasmic (TMcyto) domains (unclasped TMcyto). Conversely, in the inactive state the α/β TMcyto domains bind integrin-inactivating proteins, resulting in the association of the TMcyto domains (clasped TMcyto). Here, we report the isolation of integrin cytoplasmic tail interactors using either lipid bicelle-incorporated integrin TMcyto domains (α5, αM, αIIb, β1, β2 and β3 integrin TMcyto) or a clasped, lipid bicelle-incorporated αMβ2 TMcyto. Among the proteins found to preferentially bind clasped rather than the isolated αM and β2 subunits was L-plastin (LCP1, also known as plastin-2), which binds to and maintains the inactive state of αMβ2 integrin in vivo and thereby regulates leukocyte adhesion to integrin ligands under flow. Our findings offer a global view on cytoplasmic proteins interacting with different integrins and provide evidence for the existence of conformation-specific integrin interactors.
Collapse
Affiliation(s)
- Hui-Yuan Tseng
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Anna V Samarelli
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Patricia Kammerer
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Sarah Scholze
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Tilman Ziegler
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Roland Immler
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA.,Department of Medicine, Veterans Affairs Medical Center, Nashville, 37232 Tennessee, USA
| | - Markus Sperandio
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Charles R Sanders
- Department of Biochemistry, Center for Structural Biology, and Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, 37232 Tennessee, USA
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Ralph T Böttcher
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany .,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| |
Collapse
|
16
|
Jaykumar AB, Caceres PS, King-Medina KN, Liao TD, Datta I, Maskey D, Naggert JK, Mendez M, Beierwaltes WH, Ortiz PA. Role of Alström syndrome 1 in the regulation of blood pressure and renal function. JCI Insight 2018; 3:95076. [PMID: 30385718 DOI: 10.1172/jci.insight.95076] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/26/2018] [Indexed: 01/22/2023] Open
Abstract
Elevated blood pressure (BP) and renal dysfunction are complex traits representing major global health problems. Single nucleotide polymorphisms identified by genome-wide association studies have identified the Alström syndrome 1 (ALMS1) gene locus to render susceptibility for renal dysfunction, hypertension, and chronic kidney disease (CKD). Mutations in the ALMS1 gene in humans causes Alström syndrome, characterized by progressive metabolic alterations including hypertension and CKD. Despite compelling genetic evidence, the underlying biological mechanism by which mutations in the ALMS1 gene lead to the above-mentioned pathophysiology is not understood. We modeled this effect in a KO rat model and showed that ALMS1 genetic deletion leads to hypertension. We demonstrate that the link between ALMS1 and hypertension involves the activation of the renal Na+/K+/2Cl- cotransporter NKCC2, mediated by regulation of its endocytosis. Our findings establish a link between the genetic susceptibility to hypertension, CKD, and the expression of ALMS1 through its role in a salt-reabsorbing tubular segment of the kidney. These data point to ALMS1 as a potentially novel gene involved in BP and renal function regulation.
Collapse
Affiliation(s)
- Ankita Bachhawat Jaykumar
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Paulo S Caceres
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Keyona N King-Medina
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tang-Dong Liao
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - Indrani Datta
- Department of Public Health Sciences and.,Center for Bioinformatics, Henry Ford Health System, Detroit, Michigan, USA
| | - Dipak Maskey
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | | | - Mariela Mendez
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - William H Beierwaltes
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
17
|
Balasanyan V, Watanabe K, Dempsey WP, Lewis TL, Trinh LA, Arnold DB. Structure and Function of an Actin-Based Filter in the Proximal Axon. Cell Rep 2018; 21:2696-2705. [PMID: 29212018 DOI: 10.1016/j.celrep.2017.11.046] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/24/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022] Open
Abstract
The essential organization of microtubules within neurons has been described; however, less is known about how neuronal actin is arranged and the functional implications of its arrangement. Here, we describe, in live cells, an actin-based structure in the proximal axon that selectively prevents some proteins from entering the axon while allowing the passage of others. Concentrated patches of actin in proximal axons are present shortly after axonal specification in rat and zebrafish neurons imaged live, and they mark positions where anterogradely traveling vesicles carrying dendritic proteins halt and reverse. Patches colocalize with the ARP2/3 complex, and when ARP2/3-mediated nucleation is blocked, a dendritic protein mislocalizes to the axon. Patches are highly dynamic, with few persisting longer than 30 min. In neurons in culture and in vivo, actin appears to form a contiguous, semipermeable barrier, despite its apparently sparse distribution, preventing axonal localization of constitutively active myosin Va but not myosin VI.
Collapse
Affiliation(s)
- Varuzhan Balasanyan
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Kaori Watanabe
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - William P Dempsey
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Tommy L Lewis
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Le A Trinh
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Don B Arnold
- Department of Biology, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
18
|
Hrvatin S, Hochbaum DR, Nagy MA, Cicconet M, Robertson K, Cheadle L, Zilionis R, Ratner A, Borges-Monroy R, Klein AM, Sabatini BL, Greenberg ME. Single-cell analysis of experience-dependent transcriptomic states in the mouse visual cortex. Nat Neurosci 2018; 21:120-129. [PMID: 29230054 PMCID: PMC5742025 DOI: 10.1038/s41593-017-0029-5] [Citation(s) in RCA: 344] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022]
Abstract
Activity-dependent transcriptional responses shape cortical function. However, a comprehensive understanding of the diversity of these responses across the full range of cortical cell types, and how these changes contribute to neuronal plasticity and disease, is lacking. To investigate the breadth of transcriptional changes that occur across cell types in the mouse visual cortex after exposure to light, we applied high-throughput single-cell RNA sequencing. We identified significant and divergent transcriptional responses to stimulation in each of the 30 cell types characterized, thus revealing 611 stimulus-responsive genes. Excitatory pyramidal neurons exhibited inter- and intralaminar heterogeneity in the induction of stimulus-responsive genes. Non-neuronal cells showed clear transcriptional responses that may regulate experience-dependent changes in neurovascular coupling and myelination. Together, these results reveal the dynamic landscape of the stimulus-dependent transcriptional changes occurring across cell types in the visual cortex; these changes are probably critical for cortical function and may be sites of deregulation in developmental brain disorders.
Collapse
Affiliation(s)
- Sinisa Hrvatin
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Daniel R Hochbaum
- Society of Fellows, Harvard University, Cambridge, MA, USA
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - M Aurel Nagy
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Marcelo Cicconet
- Image and Data Analysis Core, Harvard Medical School, Boston, MA, USA
| | - Keiramarie Robertson
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Lucas Cheadle
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Rapolas Zilionis
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Vilnius University Institute of Biotechnology, Vilnius, Lithuania
| | - Alex Ratner
- ICCB-L Single Cell Core, Harvard Medical School, Boston, MA, USA
| | - Rebeca Borges-Monroy
- Program for Bioinformatics and Integrative Genomics, Graduate School of Arts and Science, Division of Medical Sciences, Harvard University, Cambridge, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
19
|
Cysteine 893 is a target of regulatory thiol modifications of GluA1 AMPA receptors. PLoS One 2017; 12:e0171489. [PMID: 28152104 PMCID: PMC5289633 DOI: 10.1371/journal.pone.0171489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/21/2017] [Indexed: 11/19/2022] Open
Abstract
Recent studies indicate that glutamatergic signaling involves, and is regulated by, thiol modifying and redox-active compounds. In this study, we examined the role of a reactive cysteine residue, Cys-893, in the cytosolic C-terminal tail of GluA1 AMPA receptor as a potential regulatory target. Elimination of the thiol function by substitution of serine for Cys-893 led to increased steady-state expression level and strongly reduced interaction with SAP97, a major cytosolic interaction partner of GluA1 C-terminus. Moreover, we found that of the three cysteine residues in GluA1 C-terminal tail, Cys-893 is the predominant target for S-nitrosylation induced by exogenous nitric oxide donors in cultured cells and lysates. Co-precipitation experiments provided evidence for native association of SAP97 with neuronal nitric oxide synthase (nNOS) and for the potential coupling of Ca2+-permeable GluA1 receptors with nNOS via SAP97. Our results show that Cys-893 can serve as a molecular target for regulatory thiol modifications of GluA1 receptors, including the effects of nitric oxide.
Collapse
|
20
|
Liao KA, González-Morales N, Schöck F. Zasp52, a Core Z-disc Protein in Drosophila Indirect Flight Muscles, Interacts with α-Actinin via an Extended PDZ Domain. PLoS Genet 2016; 12:e1006400. [PMID: 27783625 PMCID: PMC5081203 DOI: 10.1371/journal.pgen.1006400] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/04/2016] [Indexed: 11/18/2022] Open
Abstract
Z-discs are organizing centers that establish and maintain myofibril structure and function. Important Z-disc proteins are α-actinin, which cross-links actin thin filaments at the Z-disc and Zasp PDZ domain proteins, which directly interact with α-actinin. Here we investigate the biochemical and genetic nature of this interaction in more detail. Zasp52 is the major Drosophila Zasp PDZ domain protein, and is required for myofibril assembly and maintenance. We show by in vitro biochemistry that the PDZ domain plus a C-terminal extension is the only area of Zasp52 involved in the interaction with α-actinin. In addition, site-directed mutagenesis of 5 amino acid residues in the N-terminal part of the PDZ domain, within the PWGFRL motif, abolish binding to α-actinin, demonstrating the importance of this motif for α-actinin binding. Rescue assays of a novel Zasp52 allele demonstrate the crucial importance of the PDZ domain for Zasp52 function. Flight assays also show that a Zasp52 mutant suppresses the α-actinin mutant phenotype, indicating that both proteins are core structural Z-disc proteins required for optimal Z-disc function. Although Zasp PDZ domain proteins are known to bind α-actinin and play a role in muscle assembly and maintenance, the details and importance of this interaction have not been assessed. Here we demonstrate that a conserved motif in the N-terminal part of the Zasp52 PDZ domain is responsible for α-actinin binding and that a C-terminal extension of the PDZ domain is required for optimal α-actinin binding. We show using transgenic animals that in the absence of the PDZ domain no aspect of myofibril assembly can be rescued. Intriguingly, α-actinin/+ heterozygous animals show irregularities in wing beat frequency, which can be suppressed by removing one copy of Zasp52. This suggests that both proteins are required at fixed levels at the Z-disc to support optimal functionality.
Collapse
Affiliation(s)
- Kuo An Liao
- Department of Biology, McGill University, 1205 Dr Penfield Avenue, Montreal, Quebec, CANADA
| | | | - Frieder Schöck
- Department of Biology, McGill University, 1205 Dr Penfield Avenue, Montreal, Quebec, CANADA
- * E-mail:
| |
Collapse
|
21
|
CPG2 Recruits Endophilin B2 to the Cytoskeleton for Activity-Dependent Endocytosis of Synaptic Glutamate Receptors. Curr Biol 2016; 26:296-308. [PMID: 26776730 DOI: 10.1016/j.cub.2015.11.071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 11/23/2022]
Abstract
Internalization of glutamate receptors at the postsynaptic membrane via clathrin-mediated endocytosis (CME) is a key mechanism for regulating synaptic strength. A role for the F-actin cytoskeleton in CME is well established, and recently, PKA-dependent association of candidate plasticity gene 2 (CPG2) with the spine-cytoskeleton has been shown to mediate synaptic glutamate receptor internalization. Yet, how the endocytic machinery is physically coupled to the actin cytoskeleton to facilitate glutamate receptor internalization has not been demonstrated. Moreover, there has been no distinction of endocytic-machinery components that are specific to activity-dependent versus constitutive glutamate receptor internalization. Here, we show that CPG2, through a direct physical interaction, recruits endophilin B2 (EndoB2) to F-actin, thus anchoring the endocytic machinery to the spine cytoskeleton and facilitating glutamate receptor internalization. Regulation of CPG2 binding to the actin cytoskeleton by protein kinase A directly impacts recruitment of EndoB2 and clathrin. Specific disruption of EndoB2 or the CPG2-EndoB2 interaction impairs activity-dependent, but not constitutive, internalization of both NMDA- and AMPA-type glutamate receptors. These results demonstrate that, through direct interactions with F-actin and EndoB2, CPG2 physically bridges the spine cytoskeleton and the endocytic machinery, and this tripartite association is critical specifically for activity-dependent CME of synaptic glutamate receptors.
Collapse
|
22
|
PDLIM1 inhibits NF-κB-mediated inflammatory signaling by sequestering the p65 subunit of NF-κB in the cytoplasm. Sci Rep 2015; 5:18327. [PMID: 26679095 PMCID: PMC4683373 DOI: 10.1038/srep18327] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/16/2015] [Indexed: 12/03/2022] Open
Abstract
Understanding the regulatory mechanisms for the NF-κB transcription factor is key to control inflammation. IκBα maintains NF-κB in an inactive form in the cytoplasm of unstimulated cells, whereas nuclear NF-κB in activated cells is degraded by PDLIM2, a nuclear ubiquitin E3 ligase that belongs to a LIM protein family. How NF-κB activation is negatively controlled, however, is not completely understood. Here we show that PDLIM1, another member of LIM proteins, negatively regulates NF-κB-mediated signaling in the cytoplasm. PDLIM1 sequestered p65 subunit of NF-κB in the cytoplasm and suppressed its nuclear translocation in an IκBα-independent, but α-actinin-4-dependent manner. Consistently, PDLIM1 deficiency lead to increased levels of nuclear p65 protein, and thus enhanced proinflammatory cytokine production in response to innate stimuli. These studies reveal an essential role of PDLIM1 in suppressing NF-κB activation and suggest that LIM proteins comprise a new family of negative regulators of NF-κB signaling through different mechanisms.
Collapse
|
23
|
Wesseling H, Want EJ, Guest PC, Rahmoune H, Holmes E, Bahn S. Hippocampal Proteomic and Metabonomic Abnormalities in Neurotransmission, Oxidative Stress, and Apoptotic Pathways in a Chronic Phencyclidine Rat Model. J Proteome Res 2015; 14:3174-87. [PMID: 26043028 DOI: 10.1021/acs.jproteome.5b00105] [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] [Indexed: 01/27/2023]
Abstract
Schizophrenia is a neuropsychiatric disorder affecting 1% of the world's population. Due to both a broad range of symptoms and disease heterogeneity, current therapeutic approaches to treat schizophrenia fail to address all symptomatic manifestations of the disease. Therefore, disease models that reproduce core pathological features of schizophrenia are needed for the elucidation of pathological disease mechanisms. Here, we employ a comprehensive global label-free liquid chromatography-mass spectrometry proteomic (LC-MS(E)) and metabonomic (LC-MS) profiling analysis combined with the targeted proteomics (selected reaction monitoring and multiplex immunoassay) of serum and brain tissues to investigate a chronic phencyclidine (PCP) rat model in which glutamatergic hypofunction is induced through noncompetitive NMDAR-receptor antagonism. Using a multiplex immunoassay, we identified alterations in the levels of several cytokines (IL-5, IL-2, and IL-1β) and fibroblast growth factor-2. Extensive proteomic and metabonomic brain tissue profiling revealed a more prominent effect of chronic PCP treatment on both the hippocampal proteome and metabonome compared to the effect on the frontal cortex. Bioinformatic pathway analysis confirmed prominent abnormalities in NMDA-receptor-associated pathways in both brain regions, as well as alterations in other neurotransmitter systems such as kainate, AMPA, and GABAergic signaling in the hippocampus and in proteins associated with neurodegeneration. We further identified abundance changes in the level of the superoxide dismutase enzyme (SODC) in both the frontal cortex and hippocampus, which indicates alterations in oxidative stress and substantiates the apoptotic pathway alterations. The present study could lead to an increased understanding of how perturbed glutamate receptor signaling affects other relevant biological pathways in schizophrenia and, therefore, support drug discovery efforts for the improved treatment of patients suffering from this debilitating psychiatric disorder.
Collapse
Affiliation(s)
- Hendrik Wesseling
- †Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1QT, U.K
| | - Elizabeth J Want
- ‡Section of Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London SW7 2AZ, U.K
| | - Paul C Guest
- †Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1QT, U.K
| | - Hassan Rahmoune
- †Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1QT, U.K
| | - Elaine Holmes
- ‡Section of Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London SW7 2AZ, U.K
| | - Sabine Bahn
- †Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1QT, U.K.,§Department of Neuroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
24
|
Kalinowska M, Chávez AE, Lutzu S, Castillo PE, Bukauskas FF, Francesconi A. Actinin-4 Governs Dendritic Spine Dynamics and Promotes Their Remodeling by Metabotropic Glutamate Receptors. J Biol Chem 2015; 290:15909-20. [PMID: 25944910 DOI: 10.1074/jbc.m115.640136] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
Dendritic spines are dynamic, actin-rich protrusions in neurons that undergo remodeling during neuronal development and activity-dependent plasticity within the central nervous system. Although group 1 metabotropic glutamate receptors (mGluRs) are critical for spine remodeling under physiopathological conditions, the molecular components linking receptor activity to structural plasticity remain unknown. Here we identify a Ca(2+)-sensitive actin-binding protein, α-actinin-4, as a novel group 1 mGluR-interacting partner that orchestrates spine dynamics and morphogenesis in primary neurons. Functional silencing of α-actinin-4 abolished spine elongation and turnover stimulated by group 1 mGluRs despite intact surface receptor expression and downstream ERK1/2 signaling. This function of α-actinin-4 in spine dynamics was underscored by gain-of-function phenotypes in untreated neurons. Here α-actinin-4 induced spine head enlargement, a morphological change requiring the C-terminal domain of α-actinin-4 that binds to CaMKII, an interaction we showed to be regulated by group 1 mGluR activation. Our data provide mechanistic insights into spine remodeling by metabotropic signaling and identify α-actinin-4 as a critical effector of structural plasticity within neurons.
Collapse
Affiliation(s)
- Magdalena Kalinowska
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Andrés E Chávez
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Stefano Lutzu
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Pablo E Castillo
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Feliksas F Bukauskas
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Anna Francesconi
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
25
|
Hanley JG. Actin-dependent mechanisms in AMPA receptor trafficking. Front Cell Neurosci 2014; 8:381. [PMID: 25429259 PMCID: PMC4228833 DOI: 10.3389/fncel.2014.00381] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/24/2014] [Indexed: 11/22/2022] Open
Abstract
The precise regulation of AMPA receptor (AMPAR) number and subtype at the synapse is crucial for the regulation of excitatory neurotransmission, synaptic plasticity and the consequent formation of appropriate neural circuits for learning and memory. AMPAR trafficking involves the dynamic processes of exocytosis, endocytosis and endosomal recycling, all of which involve the actin cytoskeleton. The actin cytoskeleton is highly dynamic and highly regulated by an abundance of actin-binding proteins and upstream signaling pathways that modulate actin polymerization and depolymerization. Actin dynamics generate forces that manipulate membranes in the process of vesicle biogenesis, and also for propelling vesicles through the cytoplasm to reach their destination. In addition, trafficking mechanisms exploit more stable aspects of the actin cytoskeleton by using actin-based motor proteins to traffic vesicular cargo along actin filaments. Numerous studies have shown that actin dynamics are critical for AMPAR localization and function. The identification of actin-binding proteins that physically interact with AMPAR subunits, and research into their mode of action is starting to shed light on the mechanisms involved. Such proteins either regulate actin dynamics to modulate mechanical forces exerted on AMPAR-containing membranes, or associate with actin filaments to target or transport AMPAR-containing vesicles to specific subcellular regions. In addition, actin-regulatory proteins that do not physically interact with AMPARs may influence AMPAR trafficking by regulating the local actin environment in the dendritic spine.
Collapse
|
26
|
Lamprecht R. The actin cytoskeleton in memory formation. Prog Neurobiol 2014; 117:1-19. [DOI: 10.1016/j.pneurobio.2014.02.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/21/2023]
|
27
|
Abstract
α-Actinins are a major class of actin filament cross-linking proteins expressed in virtually all cells. In muscle, actinins cross-link thin filaments from adjacent sarcomeres. In non-muscle cells, different actinin isoforms play analogous roles in cross-linking actin filaments and anchoring them to structures such as cell-cell and cell-matrix junctions. Although actinins have long been known to play roles in cytokinesis, cell adhesion and cell migration, recent studies have provided further mechanistic insights into these functions. Roles for actinins in synaptic plasticity and membrane trafficking events have emerged more recently, as has a 'non-canonical' function for actinins in transcriptional regulation in the nucleus. In the present paper we review recent advances in our understanding of these diverse cell biological functions of actinins in non-muscle cells, as well as their roles in cancer and in genetic disorders affecting platelet and kidney physiology. We also make two proposals with regard to the actinin nomenclature. First, we argue that naming actinin isoforms according to their expression patterns is problematic and we suggest a more precise nomenclature system. Secondly, we suggest that the α in α-actinin is superfluous and can be omitted.
Collapse
|
28
|
Neumeier M, Krautbauer S, Schmidhofer S, Hader Y, Eisinger K, Eggenhofer E, Froehner SC, Adams ME, Mages W, Buechler C. Adiponectin receptor 1 C-terminus interacts with PDZ-domain proteins such as syntrophins. Exp Mol Pathol 2013; 95:180-6. [PMID: 23860432 DOI: 10.1016/j.yexmp.2013.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 01/22/2023]
Abstract
Adiponectin receptor 1 (AdipoR1) is one of the two signaling receptors of adiponectin with multiple beneficial effects in metabolic diseases. AdipoR1 C-terminal peptide is concordant with the consensus sequence of class I PSD-95, disc large, ZO-1 (PDZ) proteins, and screening of a liver yeast two hybrid library identified binding to β2-syntrophin (SNTB2). Hybridization of a PDZ-domain array with AdipoR1 C-terminal peptide shows association with PDZ-domains of further proteins including β1- and α-syntrophin (SNTA). Interaction of PDZ proteins and C-terminal peptides requires a free carboxy terminus next to the PDZ-binding region and is blocked by carboxy terminal added tags. N-terminal tagged AdipoR1 is more highly expressed than C-terminal tagged receptor suggesting that the free carboxy terminus may form a complex with PDZ proteins to regulate cellular AdipoR1 levels. The C- and N-terminal tagged AdipoR1 proteins are mainly localized in the cytoplasma. N-terminal but not C-terminal tagged AdipoR1 colocalizes with syntrophins in adiponectin incubated Huh7 cells. Adiponectin induced hepatic phosphorylation of AMPK and p38 MAPK which are targets of AdipoR1 is, however, not blocked in SNTA and SNTB2 deficient mice. Further, AdipoR1 protein is similarly abundant in the liver of knock-out and wild type mice when kept on a standard chow or a high fat diet. In summary these data suggest that AdipoR1 protein levels are regulated by so far uncharacterized class I PDZ proteins which are distinct from SNTA and SNTB2.
Collapse
Affiliation(s)
- Markus Neumeier
- Department of Internal Medicine I, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Predicting protein-protein interactions in the post synaptic density. Mol Cell Neurosci 2013; 56:128-39. [PMID: 23628905 DOI: 10.1016/j.mcn.2013.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/09/2013] [Accepted: 04/19/2013] [Indexed: 12/27/2022] Open
Abstract
The post synaptic density (PSD) is a specialization of the cytoskeleton at the synaptic junction, composed of hundreds of different proteins. Characterizing the protein components of the PSD and their interactions can help elucidate the mechanism of long-term changes in synaptic plasticity, which underlie learning and memory. Unfortunately, our knowledge of the proteome and interactome of the PSD is still partial and noisy. In this study we describe a computational framework to improve the reconstruction of the PSD network. The approach is based on learning the characteristics of PSD protein interactions from a set of trusted interactions, expanding this set with data collected from large scale repositories, and then predicting novel interaction with proteins that are suspected to reside in the PSD. Using this method we obtained thirty predicted interactions, with more than half of which having supporting evidence in the literature. We discuss in details two of these new interactions, Lrrtm1 with PSD-95 and Src with Capg. The first may take part in a mechanism underlying glutamatergic dysfunction in schizophrenia. The second suggests an alternative mechanism to regulate dendritic spines maturation.
Collapse
|
30
|
Alp/Enigma family proteins cooperate in Z-disc formation and myofibril assembly. PLoS Genet 2013; 9:e1003342. [PMID: 23505387 PMCID: PMC3591300 DOI: 10.1371/journal.pgen.1003342] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/10/2013] [Indexed: 11/19/2022] Open
Abstract
The Drosophila Alp/Enigma family protein Zasp52 localizes to myotendinous junctions and Z-discs. It is required for terminal muscle differentiation and muscle attachment. Its vertebrate ortholog ZASP/Cypher also localizes to Z-discs, interacts with α-actinin through its PDZ domain, and is involved in Z-disc maintenance. Human mutations in ZASP cause myopathies and cardiomyopathies. Here we show that Drosophila Zasp52 is one of the earliest markers of Z-disc assembly, and we use a Zasp52-GFP fusion to document myofibril assembly by live imaging. We demonstrate that Zasp52 is required for adult Z-disc stability and pupal myofibril assembly. In addition, we show that two closely related proteins, Zasp66 and the newly identified Zasp67, are also required for adult Z-disc stability and are participating with Zasp52 in Z-disc assembly resulting in more severe, synergistic myofibril defects in double mutants. Zasp52 and Zasp66 directly bind to α-actinin, and they can also form a ternary complex. Our results indicate that Alp/Enigma family members cooperate in Z-disc assembly and myofibril formation; and we propose, based on sequence analysis, a novel class of PDZ domain likely involved in α-actinin binding.
Collapse
|
31
|
Rácz B, Weinberg RJ. Microdomains in forebrain spines: an ultrastructural perspective. Mol Neurobiol 2013; 47:77-89. [PMID: 22983912 PMCID: PMC3538892 DOI: 10.1007/s12035-012-8345-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 08/27/2012] [Indexed: 12/21/2022]
Abstract
Glutamatergic axons in the mammalian forebrain terminate predominantly onto dendritic spines. Long-term changes in the efficacy of these excitatory synapses are tightly coupled to changes in spine morphology. The reorganization of the actin cytoskeleton underlying this spine "morphing" involves numerous proteins that provide the machinery needed for adaptive cytoskeletal remodeling. Here, we review recent literature addressing the chemical architecture of the spine, focusing mainly on actin-binding proteins (ABPs). Accumulating evidence suggests that ABPs are organized into functionally distinct microdomains within the spine cytoplasm. This functional compartmentalization provides a structural basis for regulation of the spinoskeleton, offering a novel window into mechanisms underlying synaptic plasticity.
Collapse
Affiliation(s)
- Bence Rácz
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, 1078, Budapest, Hungary.
| | | |
Collapse
|
32
|
Cheng J, Dong J, Cui Y, Wang L, Wu B, Zhang C. Interacting partners of AMPA-type glutamate receptors. J Mol Neurosci 2012; 48:441-7. [PMID: 22361832 DOI: 10.1007/s12031-012-9724-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 02/10/2012] [Indexed: 01/28/2023]
Abstract
Glutamate is the principal excitatory neurotransmitter in the brain. The alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionic (AMPA) receptors, as one of several types of endogenous ionotropic glutamate receptors, mediate the fast excitatory synaptic transmission that is essential for information processing and integration in the mammalian brain. Modifications of AMPA receptors are assumed to be the molecular basis underlying learning and memory, and impairments of AMPA receptors cause certain neurological diseases, including epilepsy, autism spectrum disorders, and Alzheimer's disease. Thus, extensive studies have been conducted, and these have revealed a complex protein-protein network controlling the expression, trafficking, and function of AMPA receptors in neurons. Here, we summarize the interacting partners of AMPA-type glutamate receptors and the functional implications of these interactions.
Collapse
Affiliation(s)
- Juan Cheng
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Peking University, Beijing 100871, China
| | | | | | | | | | | |
Collapse
|
33
|
Rosenhouse‐Dantsker A, Mehta D, Levitan I. Regulation of Ion Channels by Membrane Lipids. Compr Physiol 2012; 2:31-68. [DOI: 10.1002/cphy.c110001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Lamprecht R. The roles of the actin cytoskeleton in fear memory formation. Front Behav Neurosci 2011; 5:39. [PMID: 21808614 PMCID: PMC3139223 DOI: 10.3389/fnbeh.2011.00039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 07/02/2011] [Indexed: 01/08/2023] Open
Abstract
The formation and storage of fear memory is needed to adapt behavior and avoid danger during subsequent fearful events. However, fear memory may also play a significant role in stress and anxiety disorders. When fear becomes disproportionate to that necessary to cope with a given stimulus, or begins to occur in inappropriate situations, a fear or anxiety disorder exists. Thus, the study of cellular and molecular mechanisms underpinning fear memory may shed light on the formation of memory and on anxiety and stress related disorders. Evidence indicates that fear learning leads to changes in neuronal synaptic transmission and morphology in brain areas underlying fear memory formation including the amygdala and hippocampus. The actin cytoskeleton has been shown to participate in these key neuronal processes. Recent findings show that the actin cytoskeleton is needed for fear memory formation and extinction. Moreover, the actin cytoskeleton is involved in synaptic plasticity and in neuronal morphogenesis in brain areas that mediate fear memory. The actin cytoskeleton may therefore mediate between synaptic transmission during fear learning and long-term cellular alterations mandatory for fear memory formation.
Collapse
Affiliation(s)
- Raphael Lamprecht
- Faculty of Natural Sciences, Department of Neurobiology and Ethology, University of Haifa Haifa, Israel
| |
Collapse
|
35
|
Guryanova OA, Drazba JA, Frolova EI, Chumakov PM. Actin cytoskeleton remodeling by the alternatively spliced isoform of PDLIM4/RIL protein. J Biol Chem 2011; 286:26849-59. [PMID: 21636573 DOI: 10.1074/jbc.m111.241554] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RIL (product of PDLIM4 gene) is an actin-associated protein that has previously been shown to stimulate actin bundling by interacting with actin-cross-linking protein α-actinin-1 and increasing its affinity to filamentous actin. Here, we report that the alternatively spliced isoform of RIL, denoted here as RILaltCterm, functions as a dominant-negative modulator of RIL-mediated actin reorganization. RILaltCterm is regulated at the level of protein stability, and this protein isoform accumulates particularly in response to oxidative stress. We show that the alternative C-terminal segment of RILaltCterm has a disordered structure that directs the protein to rapid degradation in the core 20 S proteasomes. Such degradation is ubiquitin-independent and can be blocked by binding to NAD(P)H quinone oxidoreductase NQO1, a detoxifying enzyme induced by prolonged exposure to oxidative stress. We show that either overexpression of RILaltCterm or its stabilization by stresses counteracts the effects produced by full-length RIL on organization of actin cytoskeleton and cell motility. Taken together, the data suggest a mechanism for fine-tuning actin cytoskeleton rearrangement in response to stresses.
Collapse
Affiliation(s)
- Olga A Guryanova
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
36
|
Modulation of cell adhesion systems by prenatal nicotine exposure in limbic brain regions of adolescent female rats. Int J Neuropsychopharmacol 2011; 14:157-74. [PMID: 20196919 PMCID: PMC5575906 DOI: 10.1017/s1461145710000179] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Maternal smoking during pregnancy (MS) has long-lasting neurobehavioural effects on the offspring. Many MS-associated psychiatric disorders begin or change symptomatology during adolescence, a period of continuous development of the central nervous system. However, the underlying molecular mechanisms are largely unknown. Given that cell adhesion molecules (CAMs) modulate various neurotransmitter systems and are associated with many psychiatric disorders, we hypothesize that CAMs are altered by prenatal treatment of nicotine, the major psychoactive component in tobacco, in adolescent brains. Pregnant Sprague-Dawley rats were treated with nicotine (3 mg/kg.d) or saline via osmotic mini-pumps from gestational days 4 to 18. Female offspring at postnatal day 35 were sacrificed, and several limbic brain regions (the caudate putamen, nucleus accumbens, prefrontal cortex, and amygdala) were dissected for evaluation of gene expression using microarray and quantitative RT-PCR techniques. Various CAMs including neurexin, immunoglobulin, cadherin, and adhesion-GPCR superfamilies, and their intracellular signalling pathways were modified by gestational nicotine treatment (GN). Among the CAM-related pathways, GN has stronger effects on cytoskeleton reorganization pathways than on gene transcription pathways. These effects were highly region dependent, with the caudate putamen showing the greatest vulnerability. Given the important roles of CAMs in neuronal development and synaptic plasticity, our findings suggest that alteration of CAMs contributes to the neurobehavioural deficits associated with MS. Further, our study underscores that low doses of nicotine produce substantial and long-lasting changes in the brain, implying that nicotine replacement therapy during pregnancy may carry many of the same risks to the offspring as MS.
Collapse
|
37
|
Nestor MW, Cai X, Stone MR, Bloch RJ, Thompson SM. The actin binding domain of βI-spectrin regulates the morphological and functional dynamics of dendritic spines. PLoS One 2011; 6:e16197. [PMID: 21297961 PMCID: PMC3031527 DOI: 10.1371/journal.pone.0016197] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/07/2010] [Indexed: 01/30/2023] Open
Abstract
Actin microfilaments regulate the size, shape and mobility of dendritic spines and are in turn regulated by actin binding proteins and small GTPases. The βI isoform of spectrin, a protein that links the actin cytoskeleton to membrane proteins, is present in spines. To understand its function, we expressed its actin-binding domain (ABD) in CA1 pyramidal neurons in hippocampal slice cultures. The ABD of βI-spectrin bundled actin in principal dendrites and was concentrated in dendritic spines, where it significantly increased the size of the spine head. These effects were not observed after expression of homologous ABDs of utrophin, dystrophin, and α-actinin. Treatment of slice cultures with latrunculin-B significantly decreased spine head size and decreased actin-GFP fluorescence in cells expressing the ABD of α-actinin, but not the ABD of βI-spectrin, suggesting that its presence inhibits actin depolymerization. We also observed an increase in the area of GFP-tagged PSD-95 in the spine head and an increase in the amplitude of mEPSCs at spines expressing the ABD of βI-spectrin. The effects of the βI-spectrin ABD on spine size and mEPSC amplitude were mimicked by expressing wild-type Rac3, a small GTPase that co-immunoprecipitates specifically with βI-spectrin in extracts of cultured cortical neurons. Spine size was normal in cells co-expressing a dominant negative Rac3 construct with the βI-spectrin ABD. We suggest that βI-spectrin is a synaptic protein that can modulate both the morphological and functional dynamics of dendritic spines, perhaps via interaction with actin and Rac3.
Collapse
Affiliation(s)
- Michael W. Nestor
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Xiang Cai
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Michele R. Stone
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Robert J. Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Scott M. Thompson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
38
|
Nakagawa T. The biochemistry, ultrastructure, and subunit assembly mechanism of AMPA receptors. Mol Neurobiol 2010; 42:161-84. [PMID: 21080238 PMCID: PMC2992128 DOI: 10.1007/s12035-010-8149-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 11/02/2010] [Indexed: 12/25/2022]
Abstract
The AMPA-type ionotropic glutamate receptors (AMPA-Rs) are tetrameric ligand-gated ion channels that play crucial roles in synaptic transmission and plasticity. Our knowledge about the ultrastructure and subunit assembly mechanisms of intact AMPA-Rs was very limited. However, the new studies using single particle EM and X-ray crystallography are revealing important insights. For example, the tetrameric crystal structure of the GluA2cryst construct provided the atomic view of the intact receptor. In addition, the single particle EM structures of the subunit assembly intermediates revealed the conformational requirement for the dimer-to-tetramer transition during the maturation of AMPA-Rs. These new data in the field provide new models and interpretations. In the brain, the native AMPA-R complexes contain auxiliary subunits that influence subunit assembly, gating, and trafficking of the AMPA-Rs. Understanding the mechanisms of the auxiliary subunits will become increasingly important to precisely describe the function of AMPA-Rs in the brain. The AMPA-R proteomics studies continuously reveal a previously unexpected degree of molecular heterogeneity of the complex. Because the AMPA-Rs are important drug targets for treating various neurological and psychiatric diseases, it is likely that these new native complexes will require detailed mechanistic analysis in the future. The current ultrastructural data on the receptors and the receptor-expressing stable cell lines that were developed during the course of these studies are useful resources for high throughput drug screening and further drug designing. Moreover, we are getting closer to understanding the precise mechanisms of AMPA-R-mediated synaptic plasticity.
Collapse
Affiliation(s)
- Terunaga Nakagawa
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
39
|
Gu J, Lee CW, Fan Y, Komlos D, Tang X, Sun C, Yu K, Hartzell HC, Chen G, Bamburg JR, Zheng JQ. ADF/cofilin-mediated actin dynamics regulate AMPA receptor trafficking during synaptic plasticity. Nat Neurosci 2010; 13:1208-15. [PMID: 20835250 PMCID: PMC2947576 DOI: 10.1038/nn.2634] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/17/2010] [Indexed: 02/08/2023]
Abstract
Dendritic spines undergo actin-based growth and shrinkage during synaptic plasticity, in which the actin depolymerizing factor (ADF)/cofilin family of actin-associated proteins are important. Elevated ADF/cofilin activities often lead to reduced spine size and immature spine morphology but can also enhance synaptic potentiation in some cases. Thus, ADF/cofilin may have distinct effects on postsynaptic structure and function. We found that ADF/cofilin-mediated actin dynamics regulated AMPA receptor (AMPAR) trafficking during synaptic potentiation, which was distinct from actin's structural role in spine morphology. Specifically, elevated ADF/cofilin activity markedly enhanced surface addition of AMPARs after chemically induced long-term potentiation (LTP), whereas inhibition of ADF/cofilin abolished AMPAR addition. We found that chemically induced LTP elicited a temporal sequence of ADF/cofilin dephosphorylation and phosphorylation that underlies AMPAR trafficking and spine enlargement. These findings suggest that temporally regulated ADF/cofilin activities function in postsynaptic modifications of receptor number and spine size during synaptic plasticity.
Collapse
Affiliation(s)
- Jiaping Gu
- Department of Cell Biology, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010; 62:405-96. [PMID: 20716669 PMCID: PMC2964903 DOI: 10.1124/pr.109.002451] [Citation(s) in RCA: 2711] [Impact Index Per Article: 180.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The mammalian ionotropic glutamate receptor family encodes 18 gene products that coassemble to form ligand-gated ion channels containing an agonist recognition site, a transmembrane ion permeation pathway, and gating elements that couple agonist-induced conformational changes to the opening or closing of the permeation pore. Glutamate receptors mediate fast excitatory synaptic transmission in the central nervous system and are localized on neuronal and non-neuronal cells. These receptors regulate a broad spectrum of processes in the brain, spinal cord, retina, and peripheral nervous system. Glutamate receptors are postulated to play important roles in numerous neurological diseases and have attracted intense scrutiny. The description of glutamate receptor structure, including its transmembrane elements, reveals a complex assembly of multiple semiautonomous extracellular domains linked to a pore-forming element with striking resemblance to an inverted potassium channel. In this review we discuss International Union of Basic and Clinical Pharmacology glutamate receptor nomenclature, structure, assembly, accessory subunits, interacting proteins, gene expression and translation, post-translational modifications, agonist and antagonist pharmacology, allosteric modulation, mechanisms of gating and permeation, roles in normal physiological function, as well as the potential therapeutic use of pharmacological agents acting at glutamate receptors.
Collapse
Affiliation(s)
- Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322-3090, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lin MT, Luján R, Watanabe M, Frerking M, Maylie J, Adelman JP. Coupled activity-dependent trafficking of synaptic SK2 channels and AMPA receptors. J Neurosci 2010; 30:11726-34. [PMID: 20810893 PMCID: PMC2952431 DOI: 10.1523/jneurosci.1411-10.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 06/14/2010] [Accepted: 07/10/2010] [Indexed: 12/21/2022] Open
Abstract
Small conductance Ca(2+)-activated K(+) type 2 (SK2) channels are expressed in the postsynaptic density of CA1 neurons where they are activated by synaptically evoked Ca(2+) influx to limit the size of EPSPs and spine Ca(2+) transients. At Schaffer collateral synapses, the induction of long-term potentiation (LTP) increases the alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR)-mediated contribution to synaptic transmission and decreases the synaptic SK2 channel contribution through protein kinase A-dependent channel endocytosis. Using a combination of electrophysiology and immunoelectron microscopy in mice, the relationship between the dynamics of spine SK2 channels and AMPARs was investigated. Unlike AMPARs, synaptic SK2 channels under basal conditions do not rapidly recycle. Furthermore, SK2 channels occupy a distinct population of endosomes separate from AMPARs. However, blocking vesicular exocytosis or the delivery of synaptic GluA1-containing AMPARs during the induction of LTP blocks SK2 channel endocytosis. By approximately 2 h after the induction of LTP, synaptic SK2 channel expression and function are restored. Thus, LTP-dependent endocytosis of SK2 is coupled to LTP-dependent AMPA exocytosis, and the approximately 2 h window after the induction of LTP during which synaptic SK2 activity is absent may be important for consolidating the later phases of LTP.
Collapse
Affiliation(s)
- Mike T. Lin
- Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Rafael Luján
- Departamento de Ciencias Médicas, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo 060-8638, Japan, and
| | | | - James Maylie
- Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon 97239
| | - John P. Adelman
- Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| |
Collapse
|
42
|
Elkins JM, Gileadi C, Shrestha L, Phillips C, Wang J, Muniz JRC, Doyle DA. Unusual binding interactions in PDZ domain crystal structures help explain binding mechanisms. Protein Sci 2010; 19:731-41. [PMID: 20120020 PMCID: PMC2867013 DOI: 10.1002/pro.349] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PDZ domains most commonly bind the C-terminus of their protein targets. Typically the C-terminal four residues of the protein target are considered as the binding motif, particularly the C-terminal residue (P0) and third-last residue (P-2) that form the major contacts with the PDZ domain's "binding groove". We solved crystal structures of seven human PDZ domains, including five of the seven PDLIM family members. The structures of GRASP, PDLIM2, PDLIM5, and PDLIM7 show a binding mode with only the C-terminal P0 residue bound in the binding groove. Importantly, in some cases, the P-2 residue formed interactions outside of the binding groove, providing insight into the influence of residues remote from the binding groove on selectivity. In the GRASP structure, we observed both canonical and noncanonical binding in the two molecules present in the asymmetric unit making a direct comparison of these binding modes possible. In addition, structures of the PDZ domains from PDLIM1 and PDLIM4 also presented here allow comparison with canonical binding for the PDLIM PDZ domain family. Although influenced by crystal packing arrangements, the structures nevertheless show that changes in the positions of PDZ domain side-chains and the alpha B helix allow noncanonical binding interactions. These interactions may be indicative of intermediate states between unbound and fully bound PDZ domain and target protein. The noncanonical "perpendicular" binding observed potentially represents the general form of a kinetic intermediate. Comparison with canonical binding suggests that the rearrangement during binding involves both the PDZ domain and its ligand.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Declan A Doyle
- *Correspondence to: Declan A. Doyle, Trinity College Dublin, School of Biochemistry and Immunology, College Green, Dublin 2, Ireland. E-mail:
| |
Collapse
|
43
|
Ziane R, Huang H, Moghadaszadeh B, Beggs AH, Levesque G, Chahine M. Cell membrane expression of cardiac sodium channel Na(v)1.5 is modulated by alpha-actinin-2 interaction. Biochemistry 2010; 49:166-78. [PMID: 19943616 DOI: 10.1021/bi901086v] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiac sodium channel Na(v)1.5 plays a critical role in heart excitability and conduction. The molecular mechanism that underlies the expression of Na(v)1.5 at the cell membrane is poorly understood. Previous studies demonstrated that cytoskeleton proteins can be involved in the regulation of cell surface expression and localization of several ion channels. We performed a yeast two-hybrid screen to identify Na(v)1.5-associated proteins that may be involved in channel function and expression. We identified alpha-actinin-2 as an interacting partner of the cytoplasmic loop connecting domains III and IV of Na(v)1.5 (Na(v)1.5/LIII-IV). Co-immunoprecipitation and His(6) pull-down assays confirmed the physical association between Na(v)1.5 and alpha-actinin-2 and showed that the spectrin-like repeat domain is essential for binding of alpha-actinin-2 to Na(v)1.5. Patch-clamp studies revealed that the interaction with alpha-actinin-2 increases sodium channel density without changing their gating properties. Consistent with these findings, coexpression of alpha-actinin-2 and Na(v)1.5 in tsA201 cells led to an increase in the level of expression of Na(v)1.5 at the cell membrane as determined by cell surface biotinylation. Lastly, immunostaining experiments showed that alpha-actinin-2 was colocalized with Na(v)1.5 along the Z-lines and in the plasma membrane. Our data suggest that alpha-actinin-2, which is known to regulate the functional expression of the potassium channels, may play a role in anchoring Na(v)1.5 to the membrane by connecting the channel to the actin cytoskeleton network.
Collapse
Affiliation(s)
- Rahima Ziane
- Centre de Recherche Université Laval Robert-Giffard, Quebec City, QC, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Spine Remodeling and Synaptic Modification. Mol Neurobiol 2010; 41:29-41. [DOI: 10.1007/s12035-009-8093-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 12/09/2009] [Indexed: 01/05/2023]
|
45
|
Zheng M, Cheng H, Banerjee I, Chen J. ALP/Enigma PDZ-LIM domain proteins in the heart. J Mol Cell Biol 2009; 2:96-102. [PMID: 20042479 DOI: 10.1093/jmcb/mjp038] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Actinin-associated LIM protein (ALP) and Enigma are two subfamilies of Postsynaptic density 95, discs large and zonula occludens-1 (PDZ)-Lin-11, Isl1 and Mec-3 (LIM) domain containing proteins. ALP family members have one PDZ and one LIM domain, whereas Enigma proteins contain one PDZ and three LIM domains. Four ALP and three Enigma proteins have been identified in mammals, each having multiple splice variants and unique expression patterns. Functionally, these proteins bind through their PDZ domains to alpha-actinin and bind through their LIM domains or other internal protein interaction domains to other proteins, including signaling molecules. ALP and Enigma proteins have been implicated in cardiac and skeletal muscle structure, function and disease, neuronal function, bipolar disorder, tumor growth, platelet and epithelial cell motility and bone formation. This review will focus on recent advances in the biological roles of ALP/Enigma PDZ-LIM domain proteins in cardiac muscle and provide insights into mechanisms by which mutations in these proteins are related to human cardiac disease.
Collapse
Affiliation(s)
- Ming Zheng
- Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | | | | | | |
Collapse
|
46
|
Gisselsson L, Toresson H, Ruscher K, Wieloch T. Rho kinase inhibition protects CA1 cells in organotypic hippocampal slices during in vitro ischemia. Brain Res 2009; 1316:92-100. [PMID: 20026316 DOI: 10.1016/j.brainres.2009.11.087] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 11/23/2009] [Indexed: 01/24/2023]
Abstract
The actin cytoskeleton is a dynamic superstructure that regulates multiple cellular functions and that has been implicated in cell death regulation. We investigated whether modulating the neuronal actin cytoskeleton polymerization by Rho-GTPase kinase (ROCK) inhibition influences cell death in hippocampal neuronal cultures and in murine organotypic hippocampal slice cultures subjected to in vitro ischemia (IVI). During IVI, spines on vehicle treated hippocampal neurons collapsed and large dendritic actin aggregates were formed. Following ROCK inhibition by Y27632, the actin aggregates were markedly smaller while large filopodia extended from the dendritic trunk. Y27632 also provided strong neuroprotection of hippocampal pyramidal CA1 neurons, which was of similar magnitude as protection by NMDA receptor blockade. Likewise, treatment with the F-actin depolymerizing agent latrunculin during IVI diminished actin aggregation and mitigated cell death following IVI. We propose that ROCK inhibition protects neurons against ischemic damage by disrupting actin polymerization thereby mitigating NMDA receptor induced toxicity and releasing ATP bound to actin for cellular energy use. We conclude that ROCK inhibitors abrogate multiple detrimental processes and could therefore be useful in stroke therapy.
Collapse
Affiliation(s)
- Lennart Gisselsson
- Laboratory for Experimental Brain Research, Wallenberg Neuroscience Center, Lund University, BMC A13, S-22184 Lund, Sweden
| | | | | | | |
Collapse
|
47
|
Herrick S, Evers DM, Lee JY, Udagawa N, Pak DTS. Postsynaptic PDLIM5/Enigma Homolog binds SPAR and causes dendritic spine shrinkage. Mol Cell Neurosci 2009; 43:188-200. [PMID: 19900557 DOI: 10.1016/j.mcn.2009.10.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 10/20/2009] [Accepted: 10/29/2009] [Indexed: 12/24/2022] Open
Abstract
Dendritic spine morphology is thought to play important roles in synaptic development and plasticity, and morphological derangements in spines are correlated with several neurological disorders. Here, we identified an interaction between Spine-Associated RapGAP (SPAR), a postsynaptic protein that reorganizes actin cytoskeleton and drives dendritic spine head growth, and PDLIM5/Enigma Homolog (ENH), a PDZ-LIM (postsynaptic density-95/Discs large/zona occludens 1-Lin11/Isl-1/Mec3) family member. PDLIM5 has been implicated in susceptibility to bipolar disorder, major depression, and schizophrenia, but its function in neurological disease is poorly understood. We show that PDLIM5 is present in the postsynaptic density, where it promotes decreased dendritic spine head size and longer, filopodia-like morphology. Conversely, RNA interference against PDLIM5 or loss of PDLIM5 interaction with SPAR caused increased spine head diameter. Furthermore, PKC activation promoted delivery of PDLIM5 into dendritic spines and increased its spine colocalization with SPAR. These data reveal new postsynaptic functions for PDLIM5 in shrinkage of dendritic spines that may be relevant to its association with psychiatric illness.
Collapse
Affiliation(s)
- Scott Herrick
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | | | | | | | | |
Collapse
|
48
|
Ohno K, Kato H, Funahashi S, Hasegawa T, Sato K. Characterization of CLP36/Elfin/PDLIM1 in the nervous system. J Neurochem 2009; 111:790-800. [DOI: 10.1111/j.1471-4159.2009.06370.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
49
|
Wang J, Hu H, Wang S, Shi J, Chen S, Wei H, Xu X, Lu L. The important role of actinin-like protein (AcnA) in cytokinesis and apical dominance of hyphal cells in Aspergillus nidulans. Microbiology (Reading) 2009; 155:2714-2725. [DOI: 10.1099/mic.0.029215-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The actin cytoskeleton is involved in many processes in eukaryotic cells, including interaction with a wide variety of actin-binding proteins such as the actin-capping proteins, the actin filament nucleators and the actin cross-linking proteins. Here, we report the identification and characterization of an actinin-like protein (AcnA) from the filamentous fungus Aspergillus nidulans. Not only did the depletion of AcnA by alcA(p) promoter repression or the deletion of AcnA result in explicit abnormalities in septation and conidiation, but also the acnA mutants induced a loss of apical dominance in cells with dichotomous branching, in which a new branch was formed by splitting the existing tip in two. Consequently, the colony showed flabellate edges. Moreover, we found that the localization of the GFP–AcnA fusion was quite dynamic. In the isotropic expansion phase of the germinated spore, GFP–AcnA was organized as cortical patches with cables lining the cell wall. Subsequently, GFP–AcnA was localized to the actively growing hyphal tips and to the sites of septation in the form of combined double contractile rings. Our data suggest that AcnA plays an important role in cytokinesis and apical dominance of hyphal cells, possibly via actin-dependent polarization maintenance and medial ring establishment in A. nidulans. This is the first report, to our knowledge, of the function of an actinin-like protein in filamentous fungi.
Collapse
Affiliation(s)
- Jinjun Wang
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Hongqin Hu
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Sha Wang
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Jie Shi
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Shaochun Chen
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Hua Wei
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Xushi Xu
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| | - Ling Lu
- Nanjing Engineering and Technology Research Center for Microbiology, Jiangsu Key Laboratory for Bioresource Technology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, PR China
| |
Collapse
|
50
|
Hoe HS, Lee JY, Pak DT. Combinatorial morphogenesis of dendritic spines and filopodia by SPAR and alpha-actinin2. Biochem Biophys Res Commun 2009; 384:55-60. [PMID: 19393616 PMCID: PMC2707853 DOI: 10.1016/j.bbrc.2009.04.069] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Accepted: 04/11/2009] [Indexed: 12/20/2022]
Abstract
Rap small GTPases regulate excitatory synaptic strength and morphological plasticity of dendritic spines. Changes in spine structure are mediated by the F-actin cytoskeleton, but the link between Rap activity and actin dynamics is unclear. Here, we report a novel interaction between SPAR, a postsynaptic inhibitor of Rap, and alpha-actinin, a family of actin-cross-linking proteins. SPAR and alpha-actinin engage in bidirectional structural plasticity of dendritic spines: SPAR promotes spine head enlargement, whereas increased alpha-actinin2 expression favors dendritic spine elongation and thinning. Surprisingly, SPAR and alpha-actinin2 can function in an additive rather than antagonistic fashion at the same dendritic spine, generating combination spine/filopodia hybrids. These data identify a molecular pathway bridging the actin cytoskeleton and Rap at synapses, and suggest that formation of spines and filopodia are not necessarily opposing forms of structural plasticity.
Collapse
Affiliation(s)
- Hyang Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057-1464, USA
| | - Ji-Yun Lee
- Department of Pharmacology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057-1464, USA
| | - Daniel T.S. Pak
- Department of Pharmacology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057-1464, USA
| |
Collapse
|