1
|
Rust MB, Khudayberdiev S. Cyclase-associated protein: an actin regulator with multiple neuronal functions. Trends Cell Biol 2025; 35:278-281. [PMID: 39934054 DOI: 10.1016/j.tcb.2025.01.007] [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: 11/04/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
Studies of the past decade established cyclase-associated protein (CAP) as a key regulator of actin dynamics and associated its dysregulation with human brain disorders. However, its neuronal functions remained unknown until recent studies deciphered CAP-dependent mechanisms relevant for neuron differentiation or synapse morphogenesis, which are summarized and discussed in this forum article.
Collapse
Affiliation(s)
- Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Hans-Meerwein-Strasse 6, 35032 Marburg, Germany.
| | - Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Hans-Meerwein-Strasse 6, 35032 Marburg, Germany
| |
Collapse
|
2
|
Pelucchi S, Da Dalt L, De Cesare G, Stringhi R, D'Andrea L, La Greca F, Cambria C, Vandermeulen L, Zianni E, Musardo S, Roda S, Bonacina F, Nasini S, Lupo MG, Ferri N, Comai S, Gardoni F, Antonucci F, Scheggia D, Di Luca M, Norata GD, Marcello E. Neuronal PCSK9 regulates cognitive performances via the modulation of ApoER2 synaptic localization. Pharmacol Res 2025; 213:107652. [PMID: 39952371 DOI: 10.1016/j.phrs.2025.107652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
PCSK9 promotes the degradation of the low-density lipoprotein receptors and its inhibition by monoclonal antibodies or gene silencing approaches results in the reduction of plasma cholesterol levels coupled to that of cardiovascular events. Notably, while the liver is the primary source of circulating PCSK9, this protein is also abundantly expressed in the brain. However, its specific functions in the brain remain poorly understood. Here, we demonstrate that neuron-specific PCSK9 knockout mice exhibit impaired cognitive function, driven by alterations in hippocampal synapse morphology and synaptic plasticity mechanisms, coupled to spatial memory deficits. Among PCSK9 targets, we identified ApoER2 as the primary mediator of PCSK9-dependent effects on synaptic function. In neuronal cultures, PCSK9 downregulation affects ApoER2 synaptic membrane localization and lipid droplets abundance. In conclusion, our results highlight the critical role of neuronal PCSK9 in modulating synaptic ApoER2 and reveal the detrimental effects of its deficiency on synaptic function and cognitive performance. Our results shed light on the complex biology of PCSK9, crucial for evaluating side effects of PCSK9 inhibition and for developing new therapies targeting PCSK9 for brain disorders.
Collapse
Affiliation(s)
- Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Giulia De Cesare
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Filippo La Greca
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Clara Cambria
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Lina Vandermeulen
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Elisa Zianni
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Stefano Musardo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Silvia Roda
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Sofia Nasini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Nicola Ferri
- Department of Medicine, University of Padua, Padua, Italy; VIMM, Veneto Institute of Molecular Medicine, Padua, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Department of Biomedical Sciences, University of Padua, Padua, Italy; Department of Psychiatry, McGill University, Montreal, QC, Canada; IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy; Institute of Neuroscience, IN-CNR, Milano, Italy
| | - Diego Scheggia
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
3
|
Di Credico G, Pelucchi S, Pauli F, Stringhi R, Marcello E, Edefonti V. Nonlinear mixed-effects models to analyze actin dynamics in dendritic spines. Sci Rep 2025; 15:5790. [PMID: 39962126 PMCID: PMC11833086 DOI: 10.1038/s41598-025-87154-w] [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: 07/09/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Fluorescence recovery after photobleaching (FRAP) allows to study actin-turnover in dendritic spines by providing recovery trajectories over time within a nested data structure (i.e. spine/neuron/culture). Statistical approaches to FRAP usually consider one-phase association models to estimate recovery-curve-specific parameters and test statistical hypotheses on curve parameters either at the spine or neuron level, ignoring the nested data structure. However, this approach leads to pseudoreplication concerns. We propose a nonlinear mixed-effects model to integrate the one-phase association model estimate with the nested data structure of FRAP experiments; this also allows us to model heteroscedasticity and time dependence in the data. We used this approach to evaluate the effect of the downregulation of the actin-binding protein CAP2 on actin dynamics. Our model allows the additional modelling of the variance function across experimental conditions, which may represent a novel parameter of interest in FRAP experiments. Indeed, the detected differential effect of the experimental condition on the variance component captures the increased instability of time-specific observations around the spine-specific trajectory for the CAP2-downregulated spines compared to the control spines. We hypothesise that this parameter reflects the increased instability of the actin cytoskeleton in dendritic spines upon CAP2 downregulation. We developed an R-based Shiny application, termed FRApp, to fit the statistical models introduced without requiring programming expertise.
Collapse
Affiliation(s)
- Gioia Di Credico
- Department of Economics, Business, Mathematics and Statistics, Università degli Studi di Trieste, Trieste, Italy
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Francesco Pauli
- Department of Economics, Business, Mathematics and Statistics, Università degli Studi di Trieste, Trieste, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| | - Valeria Edefonti
- Branch of Medical Statistics, Biometry and Epidemiology 'G. A. Maccacaro', Department of Clinical Sciences and Community Health - Dipartimento di Eccellenza 2023-2027, Università degli Studi di Milano, Milan, Italy.
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
4
|
P A H, Basavaraju N, Gupta A, Kommaddi RP. Actin Cytoskeleton at the Synapse: An Alzheimer's Disease Perspective. Cytoskeleton (Hoboken) 2025. [PMID: 39840749 DOI: 10.1002/cm.21993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Actin, a ubiquitous and highly conserved cytoskeletal protein, plays a pivotal role in various cellular functions such as structural support, facilitating cell motility, and contributing to the dynamic processes of synaptic function. Apart from its established role in inducing morphological changes, recent developments in the field indicate an active involvement of actin in modulating both the structure and function of pre- and postsynaptic terminals. Within the presynapse, it is involved in the organization and trafficking of synaptic vesicles, contributing to neurotransmitter release. In the postsynapse, actin dynamically modulates dendritic spines, influencing the postsynaptic density organization and anchoring of neurotransmitter receptors. In addition, the dynamic interplay of actin at the synapse underscores its essential role in regulating neural communication. This review strives to offer a comprehensive overview of the recent advancements in understanding the multifaceted role of the actin cytoskeleton in synaptic functions. By emphasizing its aberrant regulation, we aim to provide valuable insights into the underlying mechanisms of Alzheimer's disease pathophysiology.
Collapse
Affiliation(s)
- Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Nimisha Basavaraju
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Anant Gupta
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
5
|
Padovani A, Pilotto A, Pelucchi S, D'Andrea L, Stringhi R, Gorla F, Aksan B, Caratozzolo S, Benussi A, Galli A, Tirloni C, Mauceri D, Canale A, Archetti S, Borroni B, Di Luca M, Marcello E. Cerebrospinal fluid cyclase-associated protein 2 is increased in Alzheimer's disease and correlates with tau pathology. Transl Neurodegener 2025; 14:1. [PMID: 39819731 PMCID: PMC11736935 DOI: 10.1186/s40035-024-00462-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Grants
- PRIN202039WMFP Ministero dell'Istruzione, dell'Università e della Ricerca
- 2017MYJ5TH Ministero dell'Istruzione, dell'Università e della Ricerca
- ID853981 Ministero dell'Istruzione, dell'Università e della Ricerca
- PRIN202039WMFP Ministero dell'Istruzione, dell'Università e della Ricerca
- H2020 IMI IDEA-FAST ID853981 Ministero dell'Istruzione, dell'Università e della Ricerca
- PRIN202039WMFP Ministero dell'Istruzione, dell'Università e della Ricerca
- PNRR P2022R2E8N Ministero dell'Istruzione, dell'Università e della Ricerca
- FIS00000560 - Stone Ministero dell'Istruzione, dell'Università e della Ricerca
- PNRR miss 4.2- investimento 1.3 Ministero dell'Istruzione, dell'Università e della Ricerca
- RF-2018-12366209 Ministero della Salute
- PNRR-MAD-2022-12376110 Ministero della Salute
- RF-2018-12366209 Ministero della Salute
- PNRR-MAD-2022-12376110 Ministero della Salute
- PRIN 20202THZAW Ministero della Salute
- PRIN 20202THZAW Ministero della Salute
- PNRR P2022TKN8C Ministero della Salute
- 2021-1516 Fondazione Cariplo
- AGYR2021 Life-Bio Associazione Italiana Ricerca Alzheimer
- AHA MCA Associazione Italiana Ricerca Alzheimer
- 21019 Alzheimer Forschung Initiative
- SFB1158 project A08 Deutsche Forschungsgemeinschaft
- Ministero dell’Istruzione, dell’Università e della Ricerca
Collapse
Affiliation(s)
- Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy.
- Brain Health Center, University of Brescia, Brescia, Italy.
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy.
- Neurobiorepository and Laboratory of Advanced Biological Markers, ASST Spedali Civili Hospital, University of Brescia, Brescia, Italy.
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Federica Gorla
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Salvatore Caratozzolo
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Clinic, Trieste University Hospital, Trieste, Italy
| | - Alice Galli
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Clara Tirloni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
- Department Molecular and Cellular Neuroscience, Institute of Anatomy and Cell Biology, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Antonio Canale
- Department of Statistical Sciences, University of Padova, Padua, Italy
| | | | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| |
Collapse
|
6
|
Schuldt C, Khudayberdiev S, Chandra BD, Linne U, Rust MB. Cyclase-associated protein (CAP) inhibits inverted formin 2 (INF2) to induce dendritic spine maturation. Cell Mol Life Sci 2024; 81:353. [PMID: 39154297 PMCID: PMC11335277 DOI: 10.1007/s00018-024-05393-y] [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: 03/12/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
The morphology of dendritic spines, the postsynaptic compartment of most excitatory synapses, decisively modulates the function of neuronal circuits as also evident from human brain disorders associated with altered spine density or morphology. Actin filaments (F-actin) form the backbone of spines, and a number of actin-binding proteins (ABP) have been implicated in shaping the cytoskeleton in mature spines. Instead, only little is known about the mechanisms that control the reorganization from unbranched F-actin of immature spines to the complex, highly branched cytoskeleton of mature spines. Here, we demonstrate impaired spine maturation in hippocampal neurons upon genetic inactivation of cyclase-associated protein 1 (CAP1) and CAP2, but not of CAP1 or CAP2 alone. We found a similar spine maturation defect upon overactivation of inverted formin 2 (INF2), a nucleator of unbranched F-actin with hitherto unknown synaptic function. While INF2 overactivation failed in altering spine density or morphology in CAP-deficient neurons, INF2 inactivation largely rescued their spine defects. From our data we conclude that CAPs inhibit INF2 to induce spine maturation. Since we previously showed that CAPs promote cofilin1-mediated cytoskeletal remodeling in mature spines, we identified them as a molecular switch that control transition from filopodia-like to mature spines.
Collapse
Affiliation(s)
- Cara Schuldt
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Ben-David Chandra
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
| | - Uwe Linne
- Department of Chemistry, Philipps-University Marburg, 35032, Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany.
| |
Collapse
|
7
|
Jury-Garfe N, Redding-Ochoa J, You Y, Martínez P, Karahan H, Chimal-Juárez E, Johnson TS, Zhang J, Resnick S, Kim J, Troncoso JC, Lasagna-Reeves CA. Enhanced microglial dynamics and a paucity of tau seeding in the amyloid plaque microenvironment contribute to cognitive resilience in Alzheimer's disease. Acta Neuropathol 2024; 148:15. [PMID: 39102080 PMCID: PMC11300572 DOI: 10.1007/s00401-024-02775-1] [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: 05/21/2024] [Revised: 07/09/2024] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Asymptomatic Alzheimer's disease (AsymAD) describes the status of individuals with preserved cognition but identifiable Alzheimer's disease (AD) brain pathology (i.e., beta-amyloid (Aβ) deposits, neuritic plaques, and neurofibrillary tangles) at autopsy. In this study, we investigated the postmortem brains of a cohort of AsymAD subjects to gain insight into the mechanisms underlying resilience to AD pathology and cognitive decline. Our results showed that AsymAD cases exhibit enrichment in core plaques, decreased filamentous plaque accumulation, and increased plaque-surrounding microglia. Less pathological tau aggregation in dystrophic neurites was found in AsymAD brains than in AD brains, and tau seeding activity was comparable to that in healthy brains. We used spatial transcriptomics to characterize the plaque niche further and revealed autophagy, endocytosis, and phagocytosis as the pathways associated with the genes upregulated in the AsymAD plaque niche. Furthermore, the levels of ARP2 and CAP1, which are actin-based motility proteins that participate in the dynamics of actin filaments to allow cell motility, were increased in the microglia surrounding amyloid plaques in AsymAD cases. Our findings suggest that the amyloid-plaque microenvironment in AsymAD cases is characterized by the presence of microglia with highly efficient actin-based cell motility mechanisms and decreased tau seeding compared with that in AD brains. These two mechanisms can potentially protect against the toxic cascade initiated by Aβ, preserving brain health, and slowing AD pathology progression.
Collapse
Affiliation(s)
- Nur Jury-Garfe
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Javier Redding-Ochoa
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yanwen You
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pablo Martínez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Enrique Chimal-Juárez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Travis S Johnson
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Susan Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging and National Institute of Health, Baltimore, MD, USA
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Cristian A Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
8
|
Khudayberdiev S, Weiss K, Heinze A, Colombaretti D, Trausch N, Linne U, Rust MB. The actin-binding protein CAP1 represses MRTF-SRF-dependent gene expression in mouse cerebral cortex. Sci Signal 2024; 17:eadj0032. [PMID: 38713765 DOI: 10.1126/scisignal.adj0032] [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: 05/31/2023] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
Serum response factor (SRF) is an essential transcription factor for brain development and function. Here, we explored how an SRF cofactor, the actin monomer-sensing myocardin-related transcription factor MRTF, is regulated in mouse cortical neurons. We found that MRTF-dependent SRF activity in vitro and in vivo was repressed by cyclase-associated protein CAP1. Inactivation of the actin-binding protein CAP1 reduced the amount of actin monomers in the cytoplasm, which promoted nuclear MRTF translocation and MRTF-SRF activation. This function was independent of cofilin1 and actin-depolymerizing factor, and CAP1 loss of function in cortical neurons was not compensated by endogenous CAP2. Transcriptomic and proteomic analyses of cerebral cortex lysates from wild-type and Cap1 knockout mice supported the role of CAP1 in repressing MRTF-SRF-dependent signaling in vivo. Bioinformatic analysis identified likely MRTF-SRF target genes, which aligned with the transcriptomic and proteomic results. Together with our previous studies that implicated CAP1 in axonal growth cone function as well as the morphology and plasticity of excitatory synapses, our findings establish CAP1 as a crucial actin regulator in the brain relevant for formation of neuronal networks.
Collapse
Affiliation(s)
- Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Hans-Meerwein-Strasse 6, 35032 Marburg, Germany
| | - Kerstin Weiss
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Anika Heinze
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Dalila Colombaretti
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Nathan Trausch
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Uwe Linne
- Department of Chemistry, Philipps-University Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Hans-Meerwein-Strasse 6, 35032 Marburg, Germany
| |
Collapse
|
9
|
D'Andrea L, Audano M, Pedretti S, Pelucchi S, Stringhi R, Imperato G, De Cesare G, Cambria C, Laporte MH, Zamboni N, Antonucci F, Di Luca M, Mitro N, Marcello E. Glucose-derived glutamate drives neuronal terminal differentiation in vitro. EMBO Rep 2024; 25:991-1021. [PMID: 38243137 PMCID: PMC10933318 DOI: 10.1038/s44319-023-00048-8] [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: 07/13/2023] [Revised: 12/01/2023] [Accepted: 12/19/2023] [Indexed: 01/21/2024] Open
Abstract
Neuronal maturation is the phase during which neurons acquire their final characteristics in terms of morphology, electrical activity, and metabolism. However, little is known about the metabolic pathways governing neuronal maturation. Here, we investigate the contribution of the main metabolic pathways, namely glucose, glutamine, and fatty acid oxidation, during the maturation of primary rat hippocampal neurons. Blunting glucose oxidation through the genetic and chemical inhibition of the mitochondrial pyruvate transporter reveals that this protein is critical for the production of glutamate, which is required for neuronal arborization, proper dendritic elongation, and spine formation. Glutamate supplementation in the early phase of differentiation restores morphological defects and synaptic function in mitochondrial pyruvate transporter-inhibited cells. Furthermore, the selective activation of metabotropic glutamate receptors restores the impairment of neuronal differentiation due to the reduced generation of glucose-derived glutamate and rescues synaptic local translation. Fatty acid oxidation does not impact neuronal maturation. Whereas glutamine metabolism is important for mitochondria, it is not for endogenous glutamate production. Our results provide insights into the role of glucose-derived glutamate as a key player in neuronal terminal differentiation.
Collapse
Affiliation(s)
- Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Gabriele Imperato
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Giulia De Cesare
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Clara Cambria
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Via F.lli Cervi 93, Segrate, 20054 Milan and via Vanvitelli 32, Milan, Italy
| | - Marine H Laporte
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Via F.lli Cervi 93, Segrate, 20054 Milan and via Vanvitelli 32, Milan, Italy
- Institute of Neuroscience, IN-CNR, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy.
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
10
|
Negrutskii BS, Porubleva LV, Malinowska A, Novosylna OV, Dadlez M, Knudsen CR. Understanding functions of eEF1 translation elongation factors beyond translation. A proteomic approach. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:67-99. [PMID: 38220433 DOI: 10.1016/bs.apcsb.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Mammalian translation elongation factors eEF1A1 and eEF1A2 are 92% homologous isoforms whose mutually exclusive tissue-specific expression is regulated during development. The isoforms have similar translation functionality, but show differences in spatial organization and participation in various processes, such as oncogenesis and virus reproduction. The differences may be due to their ability to interact with isoform-specific partner proteins. We used the identified sets of eEF1A1 or eEF1A2 partner proteins to identify cell complexes and/or processes specific to one particular isoform. As a result, we found isoform-specific interactions reflecting the involvement of different eEF1A isoforms in different cellular processes, including actin-related, chromatin-remodeling, ribonuclease H2, adenylyl cyclase, and Cul3-RING ubiquitin ligase complexes as well as initiation of mitochondrial transcription. An essential by-product of our analysis is the elucidation of a number of cellular processes beyond protein biosynthesis, where both isoforms appear to participate such as large ribosomal subunit biogenesis, mRNA splicing, DNA mismatch repair, 26S proteasome activity, P-body and exosomes formation, protein targeting to the membrane. This information suggests that a relatively high content of eEF1A in the cell may be necessary not only to maintain efficient translation, but also to ensure its participation in various cellular processes, where some roles of eEF1A have not yet been described. We believe that the data presented here will be useful for deciphering new auxiliary functions of eEF1A and its isoforms, and provide a new look at the known non-canonical functions of this main component of the human translation-elongation machinery.
Collapse
Affiliation(s)
- Boris S Negrutskii
- Institute of Molecular Biology and Genetics, Kyiv, Ukraine; Aarhus Institute of Advanced Sciences, Høegh-Guldbergs, Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen, Aarhus C, Denmark.
| | | | - Agata Malinowska
- Institute of Biochemistry and Biophysics, PAN, Pawinskiego, Warsaw, Poland
| | | | - Michal Dadlez
- Institute of Biochemistry and Biophysics, PAN, Pawinskiego, Warsaw, Poland
| | - Charlotte R Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen, Aarhus C, Denmark
| |
Collapse
|
11
|
Heinze A, Rust MB. Loss of the actin regulator cyclase-associated protein 1 (CAP1) modestly affects dendritic spine remodeling during synaptic plasticity. Eur J Cell Biol 2023; 102:151357. [PMID: 37634312 DOI: 10.1016/j.ejcb.2023.151357] [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: 05/16/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023] Open
Abstract
Dendritic spines form the postsynaptic compartment of most excitatory synapses in the vertebrate brain. Morphological changes of dendritic spines contribute to major forms of synaptic plasticity such as long-term potentiation (LTP) or depression (LTD). Synaptic plasticity underlies learning and memory, and defects in synaptic plasticity contribute to the pathogeneses of human brain disorders. Hence, deciphering the molecules that drive spine remodeling during synaptic plasticity is critical for understanding the neuronal basis of physiological and pathological brain function. Since actin filaments (F-actin) define dendritic spine morphology, actin-binding proteins (ABP) that accelerate dis-/assembly of F-actin moved into the focus as critical regulators of synaptic plasticity. We recently identified cyclase-associated protein 1 (CAP1) as a novel actin regulator in neurons that cooperates with cofilin1, an ABP relevant for synaptic plasticity. We therefore hypothesized a crucial role for CAP1 in structural synaptic plasticity. By exploiting mouse hippocampal neurons, we tested this hypothesis in the present study. We found that induction of both forms of synaptic plasticity oppositely altered concentration of exogenous, myc-tagged CAP1 in dendritic spines, with chemical LTP (cLTP) decreasing and chemical LTD (cLTD) increasing it. cLTP induced spine enlargement in CAP1-deficient neurons. However, it did not increase the density of large spines, different from control neurons. cLTD induced spine retraction and spine size reduction in control neurons, but not in CAP1-KO neurons. Together, we report that postsynaptic myc-CAP1 concentration oppositely changed during cLTP and cTLD and that CAP1 inactivation modestly affected structural plasticity.
Collapse
Affiliation(s)
- Anika Heinze
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
12
|
Pelucchi S, Macchi C, D'Andrea L, Rossi PD, Speciani MC, Stringhi R, Ruscica M, Arosio B, Di Luca M, Cesari M, Edefonti V, Marcello E. An association study of cyclase-associated protein 2 and frailty. Aging Cell 2023; 22:e13918. [PMID: 37537790 PMCID: PMC10497846 DOI: 10.1111/acel.13918] [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: 03/07/2023] [Revised: 06/06/2023] [Accepted: 06/17/2023] [Indexed: 08/05/2023] Open
Abstract
Frailty is a geriatric syndrome that results from multisystem impairment caused by age-associated accumulation of deficits. The frailty index is used to define the level of frailty. Several studies have searched for molecular biomarkers associated with frailty, to meet the needs for personalized care. Cyclase-associated protein 2 (CAP2) is a multifunctional actin-binding protein involved in various physiological and pathological processes, that might reflect frailty's intrinsic complexity. This study aimed to investigate the association between frailty index and circulating CAP2 concentration in 467 community-dwelling older adults (median age: 79; range: 65-92 years) from Milan, Italy. The selected robust regression model showed that circulating CAP2 concentration was not associated with chronological age, as well as sex and education. However, circulating CAP2 concentration was significantly and inversely associated with the frailty index: a 0.1-unit increase in frailty index leads to ~0.5-point mean decrease in CAP2 concentration. Furthermore, mean CAP2 concentration was significantly lower in frail participants (i.e., frailty index ≥0.25) than in non-frail participants. This study shows the association between serum CAP2 concentration and frailty status for the first time, highlighting the potential of CAP2 as a biomarker for age-associated accumulation of deficits.
Collapse
Affiliation(s)
- Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
| | - Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
| | - Paolo Dionigi Rossi
- Geriatric UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- General MedicineHospital San Leopoldo MandicMerateItaly
| | - Michela Carola Speciani
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro"Università degli Studi di MilanoMilanItaly
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFondazione IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Beatrice Arosio
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
| | - Matteo Cesari
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Valeria Edefonti
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro"Università degli Studi di MilanoMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti"Università degli Studi di MilanoMilanItaly
| |
Collapse
|
13
|
Jury-Garfe N, You Y, Martínez P, Redding-Ochoa J, Karahan H, Johnson TS, Zhang J, Kim J, Troncoso JC, Lasagna-Reeves CA. Enhanced microglial dynamics and paucity of tau seeding in the amyloid plaque microenvironment contributes to cognitive resilience in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550884. [PMID: 37546928 PMCID: PMC10402121 DOI: 10.1101/2023.07.27.550884] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Asymptomatic Alzheimer's disease (AsymAD) describes the status of subjects with preserved cognition but with identifiable Alzheimer's disease (AD) brain pathology (i.e. Aβ-amyloid deposits, neuritic plaques, and neurofibrillary tangles) at autopsy. In this study, we investigated the postmortem brains of a cohort of AsymAD cases to gain insight into the underlying mechanisms of resilience to AD pathology and cognitive decline. Our results showed that AsymAD cases exhibit an enrichment of core plaques and decreased filamentous plaque accumulation, as well as an increase in microglia surrounding this last type. In AsymAD cases we found less pathological tau aggregation in dystrophic neurites compared to AD and tau seeding activity comparable to healthy control subjects. We used spatial transcriptomics to further characterize the plaque niche and found autophagy, endocytosis, and phagocytosis within the top upregulated pathways in the AsymAD plaque niche, but not in AD. Furthermore, we found ARP2, an actin-based motility protein crucial to initiate the formation of new actin filaments, increased within microglia in the proximity of amyloid plaques in AsymAD. Our findings support that the amyloid-plaque microenvironment in AsymAD cases is characterized by microglia with highly efficient actin-based cell motility mechanisms and decreased tau seeding compared to AD. These two mechanisms can potentially provide protection against the toxic cascade initiated by Aβ that preserves brain health and slows down the progression of AD pathology.
Collapse
Affiliation(s)
- Nur Jury-Garfe
- Stark Neuroscience Research Institute, Indiana University, Indianapolis, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yanwen You
- Stark Neuroscience Research Institute, Indiana University, Indianapolis, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pablo Martínez
- Stark Neuroscience Research Institute, Indiana University, Indianapolis, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Javier Redding-Ochoa
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Hande Karahan
- Stark Neuroscience Research Institute, Indiana University, Indianapolis, USA
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Travis S. Johnson
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, USA
| | - Jie Zhang
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jungsu Kim
- Stark Neuroscience Research Institute, Indiana University, Indianapolis, USA
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Juan C. Troncoso
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Cristian A. Lasagna-Reeves
- Stark Neuroscience Research Institute, Indiana University, Indianapolis, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
14
|
Marcello E. Beyond the unfolded protein response: Disclosing the role of XBP1s in Alzheimer's disease. Mol Ther 2023; 31:1868-1869. [PMID: 37172589 PMCID: PMC10362413 DOI: 10.1016/j.ymthe.2023.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Affiliation(s)
- Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy.
| |
Collapse
|
15
|
Heinze A, Schuldt C, Khudayberdiev S, van Bommel B, Hacker D, Schulz TG, Stringhi R, Marcello E, Mikhaylova M, Rust MB. Functional interdependence of the actin regulators CAP1 and cofilin1 in control of dendritic spine morphology. Cell Mol Life Sci 2022; 79:558. [PMID: 36264429 PMCID: PMC9585016 DOI: 10.1007/s00018-022-04593-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/01/2022]
Abstract
The vast majority of excitatory synapses are formed on small dendritic protrusions termed dendritic spines. Dendritic spines vary in size and density that are crucial determinants of excitatory synaptic transmission. Aberrations in spine morphogenesis can compromise brain function and have been associated with neuropsychiatric disorders. Actin filaments (F-actin) are the major structural component of dendritic spines, and therefore, actin-binding proteins (ABP) that control F-actin dis-/assembly moved into the focus as critical regulators of brain function. Studies of the past decade identified the ABP cofilin1 as a key regulator of spine morphology, synaptic transmission, and behavior, and they emphasized the necessity for a tight control of cofilin1 to ensure proper brain function. Here, we report spine enrichment of cyclase-associated protein 1 (CAP1), a conserved multidomain protein with largely unknown physiological functions. Super-resolution microscopy and live cell imaging of CAP1-deficient hippocampal neurons revealed impaired synaptic F-actin organization and dynamics associated with alterations in spine morphology. Mechanistically, we found that CAP1 cooperates with cofilin1 in spines and that its helical folded domain is relevant for this interaction. Moreover, our data proved functional interdependence of CAP1 and cofilin1 in control of spine morphology. In summary, we identified CAP1 as a novel regulator of the postsynaptic actin cytoskeleton that is essential for synaptic cofilin1 activity.
Collapse
Affiliation(s)
- Anika Heinze
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Cara Schuldt
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Bas van Bommel
- AG Optobiology, Institute of Biology, Humboldt-University, 10115, Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195, Berlin, Germany
| | - Daniela Hacker
- AG Optobiology, Institute of Biology, Humboldt-University, 10115, Berlin, Germany
- Guest Group 'Neuronal Protein Transport', Institute for Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20251, Hamburg, Germany
| | - Toni G Schulz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133, Milan, Italy
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt-University, 10115, Berlin, Germany
- Guest Group 'Neuronal Protein Transport', Institute for Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20251, Hamburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany.
- DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032, Marburg, Germany.
| |
Collapse
|
16
|
Singh AK, Rai A, Weber A, Posern G. miRNA mediated downregulation of cyclase-associated protein 1 (CAP1) is required for myoblast fusion. Front Cell Dev Biol 2022; 10:899917. [PMID: 36246999 PMCID: PMC9562714 DOI: 10.3389/fcell.2022.899917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Myoblast fusion is essential for the formation, growth, and regeneration of skeletal muscle, but the molecular mechanisms that govern fusion and myofiber formation remain poorly understood. Past studies have shown an important role of the actin cytoskeleton and actin regulators in myoblast fusion. The Cyclase-Associated Proteins (CAP) 1 and 2 recently emerged as critical regulators of actin treadmilling in higher eukaryotes including mammals. Whilst the role of CAP2 in skeletal muscle development and function is well characterized, involvement of CAP1 in this process remains elusive. Here we report that CAP1, plays a critical role in cytoskeletal remodeling during myoblast fusion and formation of myotubes. Cap1 mRNA and protein are expressed in both murine C2C12 and human LHCN-M2 myoblasts, but their abundance decreases during myogenic differentiation. Perturbing the temporally controlled expression of CAP1 by overexpression or CRISPR-Cas9 mediated knockout impaired actin rearrangement, myoblast alignment, expression of profusion molecules, differentiation into multinucleated myotubes, and myosin heavy chain expression. Endogenous Cap1 expression is post-transcriptionally downregulated during differentiation by canonical myomiRs miR-1, miR-133, and miR-206, which have conserved binding sites at the 3′ UTR of the Cap1 mRNA. Deletion of the endogenous 3′ UTR by CRISPR-Cas9 in C2C12 cells phenocopies overexpression of CAP1 by inhibiting myotube formation. Our findings implicates Cap1 and its myomiR-mediated downregulation in the myoblast fusion process and the generation of skeletal muscle.
Collapse
Affiliation(s)
- Anurag Kumar Singh
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Internal Medicine I, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- *Correspondence: Anurag Kumar Singh, ; Guido Posern,
| | - Amrita Rai
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Anja Weber
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- *Correspondence: Anurag Kumar Singh, ; Guido Posern,
| |
Collapse
|
17
|
Wurz AI, Schulz AM, O’Bryant CT, Sharp JF, Hughes RM. Cytoskeletal dysregulation and neurodegenerative disease: Formation, monitoring, and inhibition of cofilin-actin rods. Front Cell Neurosci 2022; 16:982074. [PMID: 36212686 PMCID: PMC9535683 DOI: 10.3389/fncel.2022.982074] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The presence of atypical cytoskeletal dynamics, structures, and associated morphologies is a common theme uniting numerous diseases and developmental disorders. In particular, cytoskeletal dysregulation is a common cellular feature of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. While the numerous activators and inhibitors of dysregulation present complexities for characterizing these elements as byproducts or initiators of the disease state, it is increasingly clear that a better understanding of these anomalies is critical for advancing the state of knowledge and plan of therapeutic attack. In this review, we focus on the hallmarks of cytoskeletal dysregulation that are associated with cofilin-linked actin regulation, with a particular emphasis on the formation, monitoring, and inhibition of cofilin-actin rods. We also review actin-associated proteins other than cofilin with links to cytoskeleton-associated neurodegenerative processes, recognizing that cofilin-actin rods comprise one strand of a vast web of interactions that occur as a result of cytoskeletal dysregulation. Our aim is to present a current perspective on cytoskeletal dysregulation, connecting recent developments in our understanding with emerging strategies for biosensing and biomimicry that will help shape future directions of the field.
Collapse
Affiliation(s)
- Anna I. Wurz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Anna M. Schulz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Collin T. O’Bryant
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Josephine F. Sharp
- Department of Chemistry, Notre Dame College, South Euclid, OH, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, NC, United States
- *Correspondence: Robert M. Hughes,
| |
Collapse
|
18
|
Sungur AÖ, Zeitouny C, Gabele L, Metz I, Wöhr M, Michaelsen-Preusse K, Rust MB. Transient reduction in dendritic spine density in brain-specific profilin1 mutant mice is associated with behavioral deficits. Front Mol Neurosci 2022; 15:952782. [PMID: 35992199 PMCID: PMC9381693 DOI: 10.3389/fnmol.2022.952782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/13/2022] [Indexed: 01/16/2023] Open
Abstract
Actin filaments form the backbone of dendritic spines, the postsynaptic compartment of most excitatory synapses in the brain. Spine density changes affect brain function, and postsynaptic actin defects have been implicated in various neuropathies. It is mandatory to identify the actin regulators that control spine density. Based on previous studies, we hypothesized a role for the actin regulator profilin1 in spine formation. We report reduced hippocampal spine density in juvenile profilin1 mutant mice together with impairments in memory formation and reduced ultrasonic communication during active social behavior. Our results, therefore, underline a previously suggested function of profilin1 in controlling spine formation and behavior in juvenile mice.
Collapse
Affiliation(s)
- A. Özge Sungur
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany
- Behavioral Neuroscience, Experimental and Biological Psychology, University of Marburg, Marburg, Germany
| | - Caroline Zeitouny
- Department of Cellular Neurobiology, Technical University (TU) Braunschweig, Braunschweig, Germany
| | - Lea Gabele
- Department of Cellular Neurobiology, Technical University (TU) Braunschweig, Braunschweig, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany
- Deutsche Forschungsgemeinschaft (German Research Foundation) (DFG) Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, Graduiertenkolleg (Gradeschool) (GRK) 2213, University of Marburg, Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, University of Marburg, Marburg, Germany
- Social and Affective Neuroscience Research Group, Laboratory of Biological Psychology, Research Unit Brain and Cognition, Faculty of Psychology and Educational Sciences, Katholeike Universiteit (KU) Leuven, Leuven, Belgium
- Leuven Brain Institute, Katholeike Universiteit (KU) Leuven, Leuven, Belgium
| | - Kristin Michaelsen-Preusse
- Department of Cellular Neurobiology, Technical University (TU) Braunschweig, Braunschweig, Germany
- Kristin Michaelsen-Preusse,
| | - Marco B. Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany
- Deutsche Forschungsgemeinschaft (German Research Foundation) (DFG) Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, Graduiertenkolleg (Gradeschool) (GRK) 2213, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Marburg, Germany
- *Correspondence: Marco B. Rust,
| |
Collapse
|
19
|
ATM rules neurodevelopment and glutamatergic transmission in the hippocampus but not in the cortex. Cell Death Dis 2022; 13:616. [PMID: 35842432 PMCID: PMC9288428 DOI: 10.1038/s41419-022-05038-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Interest in the function of ataxia-telangiectasia-mutated protein (ATM) is extensively growing as evidenced by preclinical studies that continuously link ATM with new intracellular pathways. Here, we exploited Atm+/- and Atm-/- mice and demonstrate that cognitive defects are rescued by the delivery of the antidepressant Fluoxetine (Fluox). Fluox increases levels of the chloride intruder NKCC1 exclusively at hippocampal level suggesting an ATM context-specificity. A deeper investigation of synaptic composition unveils increased Gluk-1 and Gluk-5 subunit-containing kainate receptors (KARs) levels in the hippocampus, but not in the cortex, of Atm+/- and Atm-/- mice. Analysis of postsynaptic fractions and confocal studies indicates that KARs are presynaptic while in vitro and ex vivo electrophysiology that are fully active. These changes are (i) linked to KCC2 activity, as the KCC2 blockade in Atm+/- developing neurons results in reduced KARs levels and (ii) developmental regulated. Indeed, the pharmacological inhibition of ATM kinase in adults produces different changes as identified by RNA-seq investigation. Our data display how ATM affects both inhibitory and excitatory neurotransmission, extending its role to a variety of neurological and psychiatric disorders.
Collapse
|
20
|
Gerosa L, Mazzoleni S, Rusconi F, Longaretti A, Lewerissa E, Pelucchi S, Murru L, Giannelli SG, Broccoli V, Marcello E, Kasri NN, Battaglioli E, Passafaro M, Bassani S. The epilepsy-associated protein PCDH19 undergoes NMDA receptor-dependent proteolytic cleavage and regulates the expression of immediate-early genes. Cell Rep 2022; 39:110857. [PMID: 35613587 PMCID: PMC9152703 DOI: 10.1016/j.celrep.2022.110857] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 02/11/2022] [Accepted: 05/01/2022] [Indexed: 12/02/2022] Open
Abstract
Protocadherin-19 (PCDH19) is a synaptic cell-adhesion molecule encoded by X-linked PCDH19, a gene linked with epilepsy. Here, we report a synapse-to-nucleus signaling pathway through which PCDH19 bridges neuronal activity with gene expression. In particular, we describe the NMDA receptor (NMDAR)-dependent proteolytic cleavage of PCDH19, which leads to the generation of a PCDH19 C-terminal fragment (CTF) able to enter the nucleus. We demonstrate that PCDH19 CTF associates with chromatin and with the chromatin remodeler lysine-specific demethylase 1 (LSD1) and regulates expression of immediate-early genes (IEGs). Our results are consistent with a model whereby PCDH19 favors maintenance of neuronal homeostasis via negative feedback regulation of IEG expression and provide a key to interpreting PCDH19-related hyperexcitability. PCDH19 undergoes NMDAR-dependent cleavage by ADAM10 and possibly gamma secretase In the nucleus, PCDH19 C-terminal fragment (CTF) associates with the chromatin remodeler LSD1 PCDH19 CTF favors immediate-early gene (IEG) repression PCDH19 downregulation affects LSD1 splicing by NOVA1 and increases IEG expression
Collapse
Affiliation(s)
- Laura Gerosa
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy
| | - Sara Mazzoleni
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milano, Italy
| | - Francesco Rusconi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milano, Italy
| | - Alessandra Longaretti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milano, Italy
| | - Elly Lewerissa
- Donders Institute for Brain, Cognition, and Behaviour, Department of Human Genetics, Department of Human Genetics Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy
| | - Luca Murru
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Serena Gea Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Vania Broccoli
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy; Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy
| | - Nael Nadif Kasri
- Donders Institute for Brain, Cognition, and Behaviour, Department of Human Genetics, Department of Human Genetics Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Elena Battaglioli
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milano, Italy
| | - Maria Passafaro
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Silvia Bassani
- Institute of Neuroscience, CNR, 20854 Vedano al Lambro, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy.
| |
Collapse
|
21
|
Musardo S, Therin S, Pelucchi S, D'Andrea L, Stringhi R, Ribeiro A, Manca A, Balducci C, Pagano J, Sala C, Verpelli C, Grieco V, Edefonti V, Forloni G, Gardoni F, Meli G, Di Marino D, Di Luca M, Marcello E. The development of ADAM10 endocytosis inhibitors for the treatment of Alzheimer's disease. Mol Ther 2022; 30:2474-2490. [PMID: 35390543 DOI: 10.1016/j.ymthe.2022.03.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/15/2021] [Accepted: 03/31/2022] [Indexed: 10/18/2022] Open
Abstract
The development of new therapeutic avenues that target the early stages of Alzheimer's disease (AD) is urgently necessary. ADAM10 is a sheddase that is involved in dendritic spine shaping and limits the generation of amyloid-β. ADAM10 endocytosis increases in the hippocampus of AD patients, resulting in the decreased postsynaptic localization of the enzyme. To restore this altered pathway, we developed a cell-permeable peptide (PEP3) with a strong safety profile that is able to interfere with ADAM10 endocytosis, upregulating the postsynaptic localization and activity of ADAM10. After extensive validation, experiments in a relevant animal model clarified the optimal timing of the treatment window. PEP3 administration was effective for the rescue of cognitive defects in APP/PS1 mice only if administered at an early disease stage. Increased ADAM10 activity promoted synaptic plasticity, as revealed by changes in the molecular compositions of synapses and the spine morphology. Even though further studies are required to evaluate efficacy and safety issues of long-term administration of PEP3, these results provide preclinical evidence to support the therapeutic potential of PEP3 in AD.
Collapse
Affiliation(s)
- Stefano Musardo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Sebastien Therin
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Ana Ribeiro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Annalisa Manca
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Jessica Pagano
- CNR Neuroscience Institute, Via Raoul Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Carlo Sala
- CNR Neuroscience Institute, Via Raoul Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Via Raoul Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Valeria Grieco
- Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via Dell'Università 6, 26900 Lodi, Italy
| | - Valeria Edefonti
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry, and Epidemiology "G.A. Maccacaro", Universita` degli Studi di Milano, via Celoria 22, 20133 Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Giovanni Meli
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Daniele Di Marino
- Department of Life and Environmental Sciences, New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy.
| |
Collapse
|
22
|
Rust MB, Marcello E. Disease association of cyclase-associated protein (CAP): Lessons from gene-targeted mice and human genetic studies. Eur J Cell Biol 2022; 101:151207. [PMID: 35150966 DOI: 10.1016/j.ejcb.2022.151207] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 11/03/2022] Open
Abstract
Cyclase-associated protein (CAP) is an actin binding protein that has been initially described as partner of the adenylyl cyclase in yeast. In all vertebrates and some invertebrate species, two orthologs, named CAP1 and CAP2, have been described. CAP1 and CAP2 are characterized by a similar multidomain structure, but different expression patterns. Several molecular studies clarified the biological function of the different CAP domains, and they shed light onto the mechanisms underlying CAP-dependent regulation of actin treadmilling. However, CAPs are crucial elements not only for the regulation of actin dynamics, but also for signal transduction pathways. During recent years, human genetic studies and the analysis of gene-targeted mice provided important novel insights into the physiological roles of CAPs and their involvement in the pathogenesis of several diseases. In the present review, we summarize and discuss recent progress in our understanding of CAPs' physiological functions, focusing on heart, skeletal muscle and central nervous system as well as their involvement in the mechanisms controlling metabolism. Remarkably, loss of CAPs or impairment of CAPs-dependent pathways can contribute to the pathogenesis of different diseases. Overall, these studies unraveled CAPs complexity highlighting their capability to orchestrate structural and signaling pathways in the cells.
Collapse
Affiliation(s)
- Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy.
| |
Collapse
|
23
|
Analysis of mRNA and Protein Levels of CAP2, DLG1 and ADAM10 Genes in Post-Mortem Brain of Schizophrenia, Parkinson's and Alzheimer's Disease Patients. Int J Mol Sci 2022; 23:ijms23031539. [PMID: 35163460 PMCID: PMC8835961 DOI: 10.3390/ijms23031539] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia (SCZ) is a mental illness characterized by aberrant synaptic plasticity and connectivity. A large bulk of evidence suggests genetic and functional links between postsynaptic abnormalities and SCZ. Here, we performed quantitative PCR and Western blotting analysis in the dorsolateral prefrontal cortex (DLPFC) and hippocampus of SCZ patients to investigate the mRNA and protein expression of three key spine shapers: the actin-binding protein cyclase-associated protein 2 (CAP2), the sheddase a disintegrin and metalloproteinase 10 (ADAM10), and the synapse-associated protein 97 (SAP97). Our analysis of the SCZ post-mortem brain indicated increased DLG1 mRNA in DLPFC and decreased CAP2 mRNA in the hippocampus of SCZ patients, compared to non-psychiatric control subjects, while the ADAM10 transcript was unaffected. Conversely, no differences in CAP2, SAP97, and ADAM10 protein levels were detected between SCZ and control individuals in both brain regions. To assess whether DLG1 and CAP2 transcript alterations were selective for SCZ, we also measured their expression in the superior frontal gyrus of patients affected by neurodegenerative disorders, like Parkinson’s and Alzheimer’s disease. Interestingly, also in Parkinson’s disease patients, we found a selective reduction of CAP2 mRNA levels relative to controls but unaltered protein levels. Taken together, we reported for the first time altered CAP2 expression in the brain of patients with psychiatric and neurological disorders, thus suggesting that aberrant expression of this gene may contribute to synaptic dysfunction in these neuropathologies.
Collapse
|
24
|
Pelucchi S, Gardoni F, Di Luca M, Marcello E. Synaptic dysfunction in early phases of Alzheimer's Disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:417-438. [PMID: 35034752 DOI: 10.1016/b978-0-12-819410-2.00022-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The synapse is the locus of plasticity where short-term alterations in synaptic strength are converted to long-lasting memories. In addition to the presynaptic terminal and the postsynaptic compartment, a more holistic view of the synapse includes the astrocytes and the extracellular matrix to form a tetrapartite synapse. All these four elements contribute to synapse health and are crucial for synaptic plasticity events and, thereby, for learning and memory processes. Synaptic dysfunction is a common pathogenic trait of several brain disorders. In Alzheimer's Disease, the degeneration of synapses can be detected at the early stages of pathology progression before neuronal degeneration, supporting the hypothesis that synaptic failure is a major determinant of the disease. The synapse is the place where amyloid-β peptides are generated and is the target of the toxic amyloid-β oligomers. All the elements constituting the tetrapartite synapse are altered in Alzheimer's Disease and can synergistically contribute to synaptic dysfunction. Moreover, the two main hallmarks of Alzheimer's Disease, i.e., amyloid-β and tau, act in concert to cause synaptic deficits. Deciphering the mechanisms underlying synaptic dysfunction is relevant for the development of the next-generation therapeutic strategies aimed at modifying the disease progression.
Collapse
Affiliation(s)
- Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
25
|
Bamburg JR, Minamide LS, Wiggan O, Tahtamouni LH, Kuhn TB. Cofilin and Actin Dynamics: Multiple Modes of Regulation and Their Impacts in Neuronal Development and Degeneration. Cells 2021; 10:cells10102726. [PMID: 34685706 PMCID: PMC8534876 DOI: 10.3390/cells10102726] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023] Open
Abstract
Proteins of the actin depolymerizing factor (ADF)/cofilin family are ubiquitous among eukaryotes and are essential regulators of actin dynamics and function. Mammalian neurons express cofilin-1 as the major isoform, but ADF and cofilin-2 are also expressed. All isoforms bind preferentially and cooperatively along ADP-subunits in F-actin, affecting the filament helical rotation, and when either alone or when enhanced by other proteins, promotes filament severing and subunit turnover. Although self-regulating cofilin-mediated actin dynamics can drive motility without post-translational regulation, cells utilize many mechanisms to locally control cofilin, including cooperation/competition with other proteins. Newly identified post-translational modifications function with or are independent from the well-established phosphorylation of serine 3 and provide unexplored avenues for isoform specific regulation. Cofilin modulates actin transport and function in the nucleus as well as actin organization associated with mitochondrial fission and mitophagy. Under neuronal stress conditions, cofilin-saturated F-actin fragments can undergo oxidative cross-linking and bundle together to form cofilin-actin rods. Rods form in abundance within neurons around brain ischemic lesions and can be rapidly induced in neurites of most hippocampal and cortical neurons through energy depletion or glutamate-induced excitotoxicity. In ~20% of rodent hippocampal neurons, rods form more slowly in a receptor-mediated process triggered by factors intimately connected to disease-related dementias, e.g., amyloid-β in Alzheimer’s disease. This rod-inducing pathway requires a cellular prion protein, NADPH oxidase, and G-protein coupled receptors, e.g., CXCR4 and CCR5. Here, we will review many aspects of cofilin regulation and its contribution to synaptic loss and pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Correspondence: ; Tel.: +1-970-988-9120; Fax: +1-970-491-0494
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - O’Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - Lubna H. Tahtamouni
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Biology and Biotechnology, The Hashemite University, Zarqa 13115, Jordan
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Chemistry and Biochemistry, University of Alaska, Fairbanks, AK 99775, USA
| |
Collapse
|
26
|
Namme JN, Bepari AK, Takebayashi H. Cofilin Signaling in the CNS Physiology and Neurodegeneration. Int J Mol Sci 2021; 22:ijms221910727. [PMID: 34639067 PMCID: PMC8509315 DOI: 10.3390/ijms221910727] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022] Open
Abstract
All eukaryotic cells are composed of the cytoskeleton, which plays crucial roles in coordinating diverse cellular functions such as cell division, morphology, migration, macromolecular stabilization, and protein trafficking. The cytoskeleton consists of microtubules, intermediate filaments, and actin filaments. Cofilin, an actin-depolymerizing protein, is indispensable for regulating actin dynamics in the central nervous system (CNS) development and function. Cofilin activities are spatiotemporally orchestrated by numerous extra- and intra-cellular factors. Phosphorylation at Ser-3 by kinases attenuate cofilin’s actin-binding activity. In contrast, dephosphorylation at Ser-3 enhances cofilin-induced actin depolymerization. Cofilin functions are also modulated by various binding partners or reactive oxygen species. Although the mechanism of cofilin-mediated actin dynamics has been known for decades, recent research works are unveiling the profound impacts of cofilin dysregulation in neurodegenerative pathophysiology. For instance, oxidative stress-induced increase in cofilin dephosphorylation is linked to the accumulation of tau tangles and amyloid-beta plaques in Alzheimer’s disease. In Parkinson’s disease, cofilin activation by silencing its upstream kinases increases α-synuclein-fibril entry into the cell. This review describes the molecular mechanism of cofilin-mediated actin dynamics and provides an overview of cofilin’s importance in CNS physiology and pathophysiology.
Collapse
Affiliation(s)
- Jannatun Nayem Namme
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
| | - Asim Kumar Bepari
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh;
- Correspondence: (A.K.B.); (H.T.)
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- Correspondence: (A.K.B.); (H.T.)
| |
Collapse
|
27
|
Lapeña-Luzón T, Rodríguez LR, Beltran-Beltran V, Benetó N, Pallardó FV, Gonzalez-Cabo P. Cofilin and Neurodegeneration: New Functions for an Old but Gold Protein. Brain Sci 2021; 11:brainsci11070954. [PMID: 34356188 PMCID: PMC8303701 DOI: 10.3390/brainsci11070954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Cofilin is an actin-binding protein that plays a major role in the regulation of actin dynamics, an essential cellular process. This protein has emerged as a crucial molecule for functions of the nervous system including motility and guidance of the neuronal growth cone, dendritic spine organization, axonal branching, and synaptic signalling. Recently, other important functions in cell biology such as apoptosis or the control of mitochondrial function have been attributed to cofilin. Moreover, novel mechanisms of cofilin function regulation have also been described. The activity of cofilin is controlled by complex regulatory mechanisms, with phosphorylation being the most important, since the addition of a phosphate group to cofilin renders it inactive. Due to its participation in a wide variety of key processes in the cell, cofilin has been related to a great variety of pathologies, among which neurodegenerative diseases have attracted great interest. In this review, we summarized the functions of cofilin and its regulation, emphasizing how defects in these processes have been related to different neurodegenerative diseases.
Collapse
Affiliation(s)
- Tamara Lapeña-Luzón
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (T.L.-L.); (L.R.R.); (V.B.-B.); (N.B.); (F.V.P.)
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Laura R. Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (T.L.-L.); (L.R.R.); (V.B.-B.); (N.B.); (F.V.P.)
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Vicent Beltran-Beltran
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (T.L.-L.); (L.R.R.); (V.B.-B.); (N.B.); (F.V.P.)
| | - Noelia Benetó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (T.L.-L.); (L.R.R.); (V.B.-B.); (N.B.); (F.V.P.)
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Federico V. Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (T.L.-L.); (L.R.R.); (V.B.-B.); (N.B.); (F.V.P.)
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (T.L.-L.); (L.R.R.); (V.B.-B.); (N.B.); (F.V.P.)
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
- Correspondence:
| |
Collapse
|
28
|
Augustin V, Kins S. Fe65: A Scaffolding Protein of Actin Regulators. Cells 2021; 10:cells10071599. [PMID: 34202290 PMCID: PMC8304848 DOI: 10.3390/cells10071599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/19/2023] Open
Abstract
The scaffolding protein family Fe65, composed of Fe65, Fe65L1, and Fe65L2, was identified as an interaction partner of the amyloid precursor protein (APP), which plays a key function in Alzheimer’s disease. All three Fe65 family members possess three highly conserved interaction domains, forming complexes with diverse binding partners that can be assigned to different cellular functions, such as transactivation of genes in the nucleus, modulation of calcium homeostasis and lipid metabolism, and regulation of the actin cytoskeleton. In this article, we rule out putative new intracellular signaling mechanisms of the APP-interacting protein Fe65 in the regulation of actin cytoskeleton dynamics in the context of various neuronal functions, such as cell migration, neurite outgrowth, and synaptic plasticity.
Collapse
|
29
|
Functional Redundancy of Cyclase-Associated Proteins CAP1 and CAP2 in Differentiating Neurons. Cells 2021; 10:cells10061525. [PMID: 34204261 PMCID: PMC8234816 DOI: 10.3390/cells10061525] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 01/03/2023] Open
Abstract
Cyclase-associated proteins (CAPs) are evolutionary-conserved actin-binding proteins with crucial functions in regulating actin dynamics, the spatiotemporally controlled assembly and disassembly of actin filaments (F-actin). Mammals possess two family members (CAP1 and CAP2) with different expression patterns. Unlike most other tissues, both CAPs are expressed in the brain and present in hippocampal neurons. We recently reported crucial roles for CAP1 in growth cone function, neuron differentiation, and neuron connectivity in the mouse brain. Instead, CAP2 controls dendritic spine morphology and synaptic plasticity, and its dysregulation contributes to Alzheimer's disease pathology. These findings are in line with a model in which CAP1 controls important aspects during neuron differentiation, while CAP2 is relevant in differentiated neurons. We here report CAP2 expression during neuron differentiation and its enrichment in growth cones. We therefore hypothesized that CAP2 is relevant not only in excitatory synapses, but also in differentiating neurons. However, CAP2 inactivation neither impaired growth cone morphology and motility nor neuron differentiation. Moreover, CAP2 mutant mice did not display any obvious changes in brain anatomy. Hence, differently from CAP1, CAP2 was dispensable for neuron differentiation and brain development. Interestingly, overexpression of CAP2 rescued not only growth cone size in CAP1-deficient neurons, but also their morphology and differentiation. Our data provide evidence for functional redundancy of CAP1 and CAP2 in differentiating neurons, and they suggest compensatory mechanisms in single mutant neurons.
Collapse
|
30
|
Schneider F, Duong TA, Metz I, Winkelmeier J, Hübner CA, Endesfelder U, Rust MB. Mutual functional dependence of cyclase-associated protein 1 (CAP1) and cofilin1 in neuronal actin dynamics and growth cone function. Prog Neurobiol 2021; 202:102050. [PMID: 33845164 DOI: 10.1016/j.pneurobio.2021.102050] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/14/2021] [Accepted: 04/07/2021] [Indexed: 01/20/2023]
Abstract
Neuron connectivity depends on growth cones that navigate axons through the developing brain. Growth cones protrude and retract actin-rich structures to sense guidance cues. These cues control local actin dynamics and steer growth cones towards attractants and away from repellents, thereby directing axon outgrowth. Hence, actin binding proteins (ABPs) moved into the focus as critical regulators of neuron connectivity. We found cyclase-associated protein 1 (CAP1), an ABP with unknown brain function, abundant in growth cones. Super-resolution microscopy and live cell imaging combined with pharmacological approaches on hippocampal neurons from gene-targeted mice revealed a crucial role for CAP1 in actin dynamics that is critical for growth cone morphology and function. Growth cone defects in CAP1 knockout (KO) neurons compromised neuron differentiation and was associated with impaired neuron connectivity in CAP1-KO brains. Mechanistically, by rescue experiments in double KO neurons lacking CAP1 and the key actin regulator cofilin1, we demonstrated that CAP1 was essential for cofilin1 function in growth cone actin dynamics and morphology and vice versa. Together, we identified CAP1 as a novel actin regulator in growth cones that was relevant for neuron connectivity, and we demonstrated functional interdependence of CAP1 and cofilin1 in neuronal actin dynamics and growth cone function.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany; DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-University of Marburg, 35032, Marburg, Germany
| | - Thuy-An Duong
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Jannik Winkelmeier
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology and LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043, Marburg, Germany; Department of Physics, Mellon College of Science, Carnegie-Mellon University, Pittsburgh, PA, USA
| | - Christian A Hübner
- Institute of Human Genetics, University Hospital Jena, 07743, Jena, Germany
| | - Ulrike Endesfelder
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology and LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043, Marburg, Germany; Department of Physics, Mellon College of Science, Carnegie-Mellon University, Pittsburgh, PA, USA
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, 35032, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany; DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-University of Marburg, 35032, Marburg, Germany.
| |
Collapse
|
31
|
Smith LK, Babcock IW, Minamide LS, Shaw AE, Bamburg JR, Kuhn TB. Direct interaction of HIV gp120 with neuronal CXCR4 and CCR5 receptors induces cofilin-actin rod pathology via a cellular prion protein- and NOX-dependent mechanism. PLoS One 2021; 16:e0248309. [PMID: 33705493 PMCID: PMC7951892 DOI: 10.1371/journal.pone.0248309] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/23/2021] [Indexed: 01/08/2023] Open
Abstract
Nearly 50% of individuals with long-term HIV infection are affected by the onset of progressive HIV-associated neurocognitive disorders (HAND). HIV infiltrates the central nervous system (CNS) early during primary infection where it establishes persistent infection in microglia (resident macrophages) and astrocytes that in turn release inflammatory cytokines, small neurotoxic mediators, and viral proteins. While the molecular mechanisms underlying pathology in HAND remain poorly understood, synaptodendritic damage has emerged as a hallmark of HIV infection of the CNS. Here, we report that the HIV viral envelope glycoprotein gp120 induces the formation of aberrant, rod-shaped cofilin-actin inclusions (rods) in cultured mouse hippocampal neurons via a signaling pathway common to other neurodegenerative stimuli including oligomeric, soluble amyloid-β and proinflammatory cytokines. Previous studies showed that synaptic function is impaired preferentially in the distal proximity of rods within dendrites. Our studies demonstrate gp120 binding to either chemokine co-receptor CCR5 or CXCR4 is capable of inducing rod formation, and signaling through this pathway requires active NADPH oxidase presumably through the formation of superoxide (O2-) and the expression of cellular prion protein (PrPC). These findings link gp120-mediated oxidative stress to the generation of rods, which may underlie early synaptic dysfunction observed in HAND.
Collapse
Affiliation(s)
- Lisa K. Smith
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, United States of America
| | - Isaac W. Babcock
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Alisa E. Shaw
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Thomas B. Kuhn
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, United States of America
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
32
|
Di Giaimo R, Penna E, Pizzella A, Cirillo R, Perrone-Capano C, Crispino M. Cross Talk at the Cytoskeleton-Plasma Membrane Interface: Impact on Neuronal Morphology and Functions. Int J Mol Sci 2020; 21:ijms21239133. [PMID: 33266269 PMCID: PMC7730950 DOI: 10.3390/ijms21239133] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/18/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022] Open
Abstract
The cytoskeleton and its associated proteins present at the plasma membrane not only determine the cell shape but also modulate important aspects of cell physiology such as intracellular transport including secretory and endocytic pathways. Continuous remodeling of the cell structure and intense communication with extracellular environment heavily depend on interactions between cytoskeletal elements and plasma membrane. This review focuses on the plasma membrane-cytoskeleton interface in neurons, with a special emphasis on the axon and nerve endings. We discuss the interaction between the cytoskeleton and membrane mainly in two emerging topics of neurobiology: (i) production and release of extracellular vesicles and (ii) local synthesis of new proteins at the synapses upon signaling cues. Both of these events contribute to synaptic plasticity. Our review provides new insights into the physiological and pathological significance of the cytoskeleton-membrane interface in the nervous system.
Collapse
Affiliation(s)
- Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
- Correspondence: (R.D.G.); (M.C.)
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Raffaella Cirillo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, National Research Council (CNR), 80131 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
- Correspondence: (R.D.G.); (M.C.)
| |
Collapse
|
33
|
Rust MB, Khudayberdiev S, Pelucchi S, Marcello E. CAPt'n of Actin Dynamics: Recent Advances in the Molecular, Developmental and Physiological Functions of Cyclase-Associated Protein (CAP). Front Cell Dev Biol 2020; 8:586631. [PMID: 33072768 PMCID: PMC7543520 DOI: 10.3389/fcell.2020.586631] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Cyclase-associated protein (CAP) has been discovered three decades ago in budding yeast as a protein that associates with the cyclic adenosine monophosphate (cAMP)-producing adenylyl cyclase and that suppresses a hyperactive RAS2 variant. Since that time, CAP has been identified in all eukaryotic species examined and it became evident that the activity in RAS-cAMP signaling is restricted to a limited number of species. Instead, its actin binding activity is conserved among eukaryotes and actin cytoskeleton regulation emerged as its primary function. However, for many years, the molecular functions as well as the developmental and physiological relevance of CAP remained unknown. In the present article, we will compile important recent progress on its molecular functions that identified CAP as a novel key regulator of actin dynamics, i.e., the spatiotemporally controlled assembly and disassembly of actin filaments (F-actin). These studies unraveled a cooperation with ADF/Cofilin and Twinfilin in F-actin disassembly, a nucleotide exchange activity on globular actin monomers (G-actin) that is required for F-actin assembly and an inhibitory function towards the F-actin assembly factor INF2. Moreover, by focusing on selected model organisms, we will review current literature on its developmental and physiological functions, and we will present studies implicating CAP in human pathologies. Together, this review article summarizes and discusses recent achievements in understanding the molecular, developmental and physiological functions of CAP, which led this protein emerge as a novel CAPt'n of actin dynamics.
Collapse
Affiliation(s)
- Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, University of Marburg and Justus-Liebig-University Giessen, Giessen, Germany
| | - Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|