1
|
Tatzl E, Petracco G, Faimann I, Balasso M, Mooslechner AA, Bärnthaler T, Rodriguez‐Blanco G, Reichmann F. Deficiency of the Synaptic Adhesion Protein Leucine-Rich Repeat Transmembrane Protein 4 Like 1 Affects Anxiety and Aggression in Zebrafish. Acta Physiol (Oxf) 2025; 241:e70042. [PMID: 40183503 PMCID: PMC11970230 DOI: 10.1111/apha.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/23/2025] [Indexed: 04/05/2025]
Abstract
AIM Leucine-rich repeat transmembrane proteins (LRRTMs) are synaptic adhesion proteins that regulate synapse development and function. They interact transsynaptically with presynaptic binding partners to promote presynaptic differentiation. Polymorphisms of LRRTM4, one of the four members of this protein family, have been linked to multiple neuropsychiatric disorders and childhood aggression, but the underlying mechanisms and physiological function of LRRTM4 during behavior are currently unclear. METHODS To characterize the role of this gene for brain function, we combined a battery of behavioral assays with transcriptomic and metabolomic analyses, using zebrafish as a model system. RESULTS Our findings revealed that lrrtm4l1, a brain-specific zebrafish orthologue of human LRRTM4, exhibits a brain region-specific expression pattern similar to humans, with strong expression in the dorsal telencephalon, a brain area critical for regulating emotional-affective and social behavior. lrrtm4l1-/- zebrafish displayed heightened anxiety and reduced aggression, while locomotion and social behavior remained unaffected by the gene knockout. Transcriptomic analysis of the telencephalon identified over 100 differentially expressed genes between wild-type and mutant zebrafish and an enrichment of pathways related to synaptic plasticity and neuronal signaling. The brain metabolome of lrrtm4l1-/- zebrafish showed multiple alterations, particularly in the dopaminergic and adenosinergic neurotransmitter systems. CONCLUSION These findings suggest that LRRTMs may have functions beyond their established role in excitatory synapse development, such as the regulation of neurotransmission and behavior. Targeting LRRTM4 therapeutically may thus be an interesting novel approach to alleviate excessive aggression or anxiety associated with a number of neuropsychiatric conditions.
Collapse
Affiliation(s)
- Eva Tatzl
- Division of Pharmacology, Otto Loewi Research CenterMedical University of GrazGrazAustria
| | - Giulia Petracco
- Division of Pharmacology, Otto Loewi Research CenterMedical University of GrazGrazAustria
| | - Isabella Faimann
- Division of Pharmacology, Otto Loewi Research CenterMedical University of GrazGrazAustria
| | - Marco Balasso
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Agnes Anna Mooslechner
- Division of Pharmacology, Otto Loewi Research CenterMedical University of GrazGrazAustria
| | - Thomas Bärnthaler
- Division of Pharmacology, Otto Loewi Research CenterMedical University of GrazGrazAustria
| | - Giovanny Rodriguez‐Blanco
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Florian Reichmann
- Division of Pharmacology, Otto Loewi Research CenterMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| |
Collapse
|
2
|
Tambrin HM, Liu Y, Zhu K, Teng X, Toyama Y, Miao Y, Ludwig A. ARHGAP12 suppresses F-actin assembly to control epithelial tight junction mechanics and paracellular leak pathway permeability. Cell Rep 2025; 44:115511. [PMID: 40198220 DOI: 10.1016/j.celrep.2025.115511] [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: 08/24/2024] [Revised: 02/18/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
Tight junctions (TJs) control the paracellular transport of ions, solutes, and macromolecules across epithelial barriers. There is evidence that claudin-based ion transport (the pore pathway) and the paracellular transport of macromolecules (the leak pathway) are controlled independently. However, how leak pathway flux is regulated is unclear. Here, we have identified the Cdc42/Rac GTPase-activating protein ARHGAP12 as a specific activator of the leak pathway. ARHGAP12 is recruited to TJs via an interaction between its Src homology (SH3) domain and the TJ protein ZO-2 to suppress N-WASP-mediated F-actin assembly. This dampens junctional tension and promotes the paracellular transport of macromolecules without affecting ion flux. Mechanistically, we demonstrate that the ARHGAP12 tandem WW domain interacts directly with PPxR motifs in the proline-rich domain of N-WASP and thereby attenuates SH3-domain-mediated N-WASP oligomerization and Arp2/3-driven F-actin assembly. Collectively, our data indicate that branched F-actin networks regulate junctional tension to fine-tune the TJ leak pathway.
Collapse
Affiliation(s)
- Hana Maldivita Tambrin
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Yun Liu
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Kexin Zhu
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xiang Teng
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive, Singapore 117411, Singapore
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive, Singapore 117411, Singapore
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
3
|
Yang X, Yu D, Gao F, Yang J, Chen Z, Liu J, Yang X, Li L, Zhang Y, Yan C. Integrative Analysis of Morphine-Induced Differential Circular RNAs and ceRNA Networks in the Medial Prefrontal Cortex. Mol Neurobiol 2024; 61:4602-4618. [PMID: 38109006 DOI: 10.1007/s12035-023-03859-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Circular RNAs (circRNAs) are a novel type of non-coding RNAs. Despite the fact that the functional mechanisms of most circRNAs remain unknown, emerging evidence indicates that circRNAs could sponge microRNAs (miRNAs), bind to RNA binding proteins (RBP), and even be translated into protein. Recent research has demonstrated the crucial roles played by circRNAs in neuropsychiatric disorders. The medial prefrontal cortex (mPFC) is a crucial component of drug reward circuitry and exerts top-down control over cognitive functions. However, there is currently limited knowledge about the correlation between circRNAs and morphine-associated contextual memory in the mPFC. Here, we performed morphine-induced conditioned place preference (CPP) in mice and extracted mPFC tissue for RNA-sequencing. Our study represented the first attempt to identify differentially expressed circRNAs (DEcircRNAs) and mRNAs (DEmRNAs) in the mPFC after morphine-induced CPP. We identified 47 significantly up-regulated DEcircRNAs and 429 significantly up-regulated DEmRNAs, along with 74 significantly down-regulated DEcircRNAs and 391 significantly down-regulated DEmRNAs. Functional analysis revealed that both DEcircRNAs and DEmRNAs were closely associated with neuroplasticity. To further validate the DEcircRNAs, we conducted qRT-PCR, Sanger sequencing, and RNase R digestion assays. Additionally, using an integrated bioinformatics approach, we constructed ceRNA networks and identified critical circRNA/miRNA/mRNA axes that contributed to the development of morphine-associated contextual memory. In summary, our study provided novel insights into the role of circRNAs in drug-related memory, specifically from the perspective of ceRNAs.
Collapse
Affiliation(s)
- Xixi Yang
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Dongyu Yu
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Feifei Gao
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Jingsi Yang
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Zhennan Chen
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Junlin Liu
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Xiaoyu Yang
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Lanjiang Li
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China
| | - Yuxiang Zhang
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China.
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China.
| | - Chunxia Yan
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
- Key Laboratory of Forensic Medicine, National Health Commission, Xi'an 710061, Shaanxi, China.
- Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an 710100, Shaanxi, China.
| |
Collapse
|
4
|
Fei H, Shi X, Sun D, Yang H, Wang D, Li K, Si X, Hu W. Integrated analysis identified the role of three family members of ARHGAP in pancreatic adenocarcinoma. Sci Rep 2024; 14:11790. [PMID: 38783033 PMCID: PMC11116390 DOI: 10.1038/s41598-024-62577-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
The Rho GTPase activating protein family (ARHGAPs) is expressed in pancreatic adenocarcinoma (PAAD) but its function is unclear. The aim of this study was to explore the role and potential clinical value of ARHGAPs in PAAD. Using TCGA and GEO databases to analyze expression of ARHGAPs in PAAD and normal tissues. Survival curve was drawn by Kaplan-Meier. ARHGAPs were integrated analyzed by GEPIA2, TIMER, UCLCAN, cBioPortal and R language. Protein level and prognostic value were evaluated via IHC staining or survival analysis. We totally identify 18 differentially expressed (DE) ARHGAPs in PAAD. Among the 18 DE genes, 8 were positively correlated with tumor grade; abnorrmal expression of 5 was positively correlated with copy number variation; expression of 4 was positively correlated with promoter hypomethylation. Multivariate Cox regression identified ARHGAP5, ARHGAP11A, and ARHGAP12 as independent prognostic factors of PAAD. The function of ARHGAPs was mainly related to GTPase activity and signaling, axon guidance, proteoglycans in cancer and focal adhesion. Expression of 7 ARHGAPs was strongly correlated with immune infiltration. Immunohistochemistry showed increased protein levels of ARHGAP5, ARHGAP11A, and ARHGAP12 in PAAD tissues. Survival analysis confirmed a negative correlation between ARHGAP5, ARHGAP11A, and ARHGAP12 expression and patient prognosis. Multivariate Cox regression proved ARHGAP5, ARHGAP11A, and ARHGAP12 could serve as independent prognostic indicators for PAAD. Finally, this study verified ARHGAP5, ARHGAP11A, and ARHGAP12 as independent prognostic factors in PAAD, suggesting their significance for the diagnosis and treatment of PAAD.
Collapse
Affiliation(s)
- Haoran Fei
- Department of Hepatobiliary Surgery, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu, China
- Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xiao Shi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, Jiangsu, China
| | - Dan Sun
- Department of Hepatobiliary Surgery, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu, China
- Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Haishen Yang
- Department of Hepatobiliary Surgery, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu, China
| | - Dali Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu, China
| | - Kai Li
- Department of Hepatobiliary Surgery, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu, China
| | - Xinxin Si
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, Jiangsu, China.
| | - Wei Hu
- Department of Hepatobiliary Surgery, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu, China.
| |
Collapse
|
5
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
6
|
Farini D, Cesari E, Weatheritt RJ, La Sala G, Naro C, Pagliarini V, Bonvissuto D, Medici V, Guerra M, Di Pietro C, Rizzo FR, Musella A, Carola V, Centonze D, Blencowe BJ, Marazziti D, Sette C. A Dynamic Splicing Program Ensures Proper Synaptic Connections in the Developing Cerebellum. Cell Rep 2021; 31:107703. [PMID: 32492419 DOI: 10.1016/j.celrep.2020.107703] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/13/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Tight coordination of gene expression in the developing cerebellum is crucial for establishment of neuronal circuits governing motor and cognitive function. However, transcriptional changes alone do not explain all of the switches underlying neuronal differentiation. Here we unveiled a widespread and highly dynamic splicing program that affects synaptic genes in cerebellar neurons. The motifs enriched in modulated exons implicated the splicing factor Sam68 as a regulator of this program. Sam68 controls splicing of exons with weak branchpoints by directly binding near the 3' splice site and competing with U2AF recruitment. Ablation of Sam68 disrupts splicing regulation of synaptic genes associated with neurodevelopmental diseases and impairs synaptic connections and firing of Purkinje cells, resulting in motor coordination defects, ataxia, and abnormal social behavior. These findings uncover an unexpectedly dynamic splicing regulatory network that shapes the synapse in early life and establishes motor and cognitive circuitry in the developing cerebellum.
Collapse
Affiliation(s)
- Donatella Farini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Eleonora Cesari
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Robert J Weatheritt
- Donnelly Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; EMBL Australia, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Gina La Sala
- Institute of Cell Biology and Neurobiology, CNR, Monterotondo, Rome, Italy
| | - Chiara Naro
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Vittoria Pagliarini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Davide Bonvissuto
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
| | - Vanessa Medici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Marika Guerra
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Chiara Di Pietro
- Institute of Cell Biology and Neurobiology, CNR, Monterotondo, Rome, Italy
| | - Francesca Romana Rizzo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; San Raffaele Pisana and University San Raffaele, IRCCS, Rome, Italy
| | | | - Valeria Carola
- Fondazione Santa Lucia, IRCCS, Rome, Italy; Department of Dynamic and Clinical Psychology, University of Rome Sapienza, Rome, Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Benjamin J Blencowe
- Donnelly Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Daniela Marazziti
- Institute of Cell Biology and Neurobiology, CNR, Monterotondo, Rome, Italy
| | - Claudio Sette
- Fondazione Santa Lucia, IRCCS, Rome, Italy; Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
7
|
Ji B, Skup M. Roles of palmitoylation in structural long-term synaptic plasticity. Mol Brain 2021; 14:8. [PMID: 33430908 PMCID: PMC7802216 DOI: 10.1186/s13041-020-00717-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are important cellular mechanisms underlying learning and memory processes. N-Methyl-d-aspartate receptor (NMDAR)-dependent LTP and LTD play especially crucial roles in these functions, and their expression depends on changes in the number and single channel conductance of the major ionotropic glutamate receptor α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) located on the postsynaptic membrane. Structural changes in dendritic spines comprise the morphological platform and support for molecular changes in the execution of synaptic plasticity and memory storage. At the molecular level, spine morphology is directly determined by actin cytoskeleton organization within the spine and indirectly stabilized and consolidated by scaffold proteins at the spine head. Palmitoylation, as a uniquely reversible lipid modification with the ability to regulate protein membrane localization and trafficking, plays significant roles in the structural and functional regulation of LTP and LTD. Altered structural plasticity of dendritic spines is also considered a hallmark of neurodevelopmental disorders, while genetic evidence strongly links abnormal brain function to impaired palmitoylation. Numerous studies have indicated that palmitoylation contributes to morphological spine modifications. In this review, we have gathered data showing that the regulatory proteins that modulate the actin network and scaffold proteins related to AMPAR-mediated neurotransmission also undergo palmitoylation and play roles in modifying spine architecture during structural plasticity.
Collapse
Affiliation(s)
- Benjun Ji
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Małgorzata Skup
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| |
Collapse
|
8
|
Hu J, Liu PL, Hua Y, Gao BY, Wang YY, Bai YL, Chen C. Constraint-induced movement therapy enhances AMPA receptor-dependent synaptic plasticity in the ipsilateral hemisphere following ischemic stroke. Neural Regen Res 2021; 16:319-324. [PMID: 32859791 PMCID: PMC7896237 DOI: 10.4103/1673-5374.290900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Constraint-induced movement therapy (CIMT) can promote the recovery of motor function in injured upper limbs following stroke, which may be associated with upregulation of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) at synapses in the ipsilateral sensorimotor cortex in our previous study. However, AMPAR distribution is tightly regulated, and only AMPARs on the postsynaptic membrane can mediate synaptic transmission. We speculated that synaptic remodeling induced by movement-associated synaptic activity can promote functional recovery from stroke. To test this hypothesis, we compared AMPAR expression on the postsynaptic membrane surface in a rat model of ischemic stroke induced by middle cerebral artery occlusion (MCAO) with versus without CIMT, which consisted of daily running wheel training for 2 weeks starting on day 7 after MCAO. The results showed that CIMT increased the number of glutamate receptor (GluR)2-containing functional synapses in the ipsilateral sensorimotor cortex, and reduced non-GluR2 AMPARs in the ipsilateral sensorimotor cortex and hippocampal CA3 region. In addition, CIMT enhanced AMPAR expression on the surface of post-synaptic membrane in the ipsilateral sensorimotor cortex and hippocampus. Thus, CIMT promotes the recovery of motor function of injured upper limbs following stroke by enhancing AMPAR-mediated synaptic transmission in the ischemic hemisphere. These findings provide supporting evidence for the clinical value of CIMT for restoring limb movement in stroke patients. All experimental procedures and protocols were approved by the Department of Laboratory Animal Science of Fudan University, China (approval No. 201802173S) on March 3, 2018.
Collapse
Affiliation(s)
- Jian Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Pei-Le Liu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Bei-Yao Gao
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Yuan Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Long Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chan Chen
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Wang X, Cheng Z, Xu J, Feng M, Zhang H, Zhang L, Qian L. Circular RNA Arhgap12 modulates doxorubicin-induced cardiotoxicity by sponging miR-135a-5p. Life Sci 2020; 265:118788. [PMID: 33245966 DOI: 10.1016/j.lfs.2020.118788] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/06/2020] [Accepted: 11/14/2020] [Indexed: 02/06/2023]
Abstract
AIM This study aimed to investigate the regulatory role of differentially-expressed circular RNAs (circRNAs) in mouse cardiomyocytes during doxorubicin (DOX)-induced cardiotoxicity. MAIN METHODS Two groups of mice were injected with equal volumes (0.1 mL) of normal saline and DOX. Mouse heart tissue was isolated and digested for total RNA extraction and then subjected to next-generation RNA-sequencing. Expression profiles of circRNAs and circRNA-miRNA-mRNA networks were also constructed. Overall, 48 upregulated and 16 downregulated circRNAs were found to be statistically significant (p < 0.05) in the DOX-injected group. Bioinformatics analysis revealed several potential biological pathways that might be related to apoptosis caused by DOX-induced cardiotoxicity. In addition, using qRT-PCR, we found that a circRNA coded by the Arhgap12 gene, termed circArhgap12, was upregulated in the mouse heart tissue upon DOX intervention. CircArhgap12 enhanced apoptotic cell rate, as assessed using terminal-deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay, and increased reactive oxygen species and malondialdehyde release as well as superoxide dismutase and caspase-3 activation. Using a luciferase reporter assay, we found that circArhgap12 could sponge miR-135a-5p. In rat primary cardiomyocytes, we found that si-circArhgap12 promoted apoptosis and oxidative stress by sponging the miR-135a-5p inhibitor. Using bioinformatics analysis and luciferase reporter assay, we found that miR-135a-5p might have a potential target site for ADCY1 mRNA. KEY FINDINGS Our research demonstrated that the expression profile of circRNAs was modified significantly and that circArhgap12 might play a competitive role among endogenous RNAs in mouse cardiomyocytes during DOX-induced cardiotoxicity. SIGNIFICANCE Our study may provide a preliminary understanding of DOX-induced cardiotoxicity modulated by circRNA and its competing endogenous RNAs network.
Collapse
Affiliation(s)
- Xuejun Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zijie Cheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jia Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengwen Feng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Lingmei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Department of General Medicine, Tongren Hospital of Shanghai Jiao Tong University of Medicine, Shanghai 200336, China.
| |
Collapse
|
10
|
Fedoryshchak RO, Přechová M, Butler AM, Lee R, O'Reilly N, Flynn HR, Snijders AP, Eder N, Ultanir S, Mouilleron S, Treisman R. Molecular basis for substrate specificity of the Phactr1/PP1 phosphatase holoenzyme. eLife 2020; 9:61509. [PMID: 32975518 PMCID: PMC7599070 DOI: 10.7554/elife.61509] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/24/2020] [Indexed: 01/01/2023] Open
Abstract
PPP-family phosphatases such as PP1 have little intrinsic specificity. Cofactors can target PP1 to substrates or subcellular locations, but it remains unclear how they might confer sequence-specificity on PP1. The cytoskeletal regulator Phactr1 is a neuronally enriched PP1 cofactor that is controlled by G-actin. Structural analysis showed that Phactr1 binding remodels PP1's hydrophobic groove, creating a new composite surface adjacent to the catalytic site. Using phosphoproteomics, we identified mouse fibroblast and neuronal Phactr1/PP1 substrates, which include cytoskeletal components and regulators. We determined high-resolution structures of Phactr1/PP1 bound to the dephosphorylated forms of its substrates IRSp53 and spectrin αII. Inversion of the phosphate in these holoenzyme-product complexes supports the proposed PPP-family catalytic mechanism. Substrate sequences C-terminal to the dephosphorylation site make intimate contacts with the composite Phactr1/PP1 surface, which are required for efficient dephosphorylation. Sequence specificity explains why Phactr1/PP1 exhibits orders-of-magnitude enhanced reactivity towards its substrates, compared to apo-PP1 or other PP1 holoenzymes.
Collapse
Affiliation(s)
- Roman O Fedoryshchak
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Magdalena Přechová
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Abbey M Butler
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom.,Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Rebecca Lee
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom.,Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Nicola O'Reilly
- Peptide Chemistry Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Helen R Flynn
- Proteomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Ambrosius P Snijders
- Proteomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Noreen Eder
- Proteomics Science Technology Platform, The Francis Crick Institute, London, United Kingdom.,Kinases and Brain Development Laboratory The Francis Crick Institute, London, United Kingdom
| | - Sila Ultanir
- Kinases and Brain Development Laboratory The Francis Crick Institute, London, United Kingdom
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Richard Treisman
- Signalling and Transcription Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
11
|
Diring J, Mouilleron S, McDonald NQ, Treisman R. RPEL-family rhoGAPs link Rac/Cdc42 GTP loading to G-actin availability. Nat Cell Biol 2019; 21:845-855. [PMID: 31209295 PMCID: PMC6960015 DOI: 10.1038/s41556-019-0337-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/29/2019] [Indexed: 12/29/2022]
Abstract
RPEL proteins, which contain the G-actin-binding RPEL motif, coordinate cytoskeletal processes with actin dynamics. We show that the ArhGAP12- and ArhGAP32-family GTPase-activating proteins (GAPs) are RPEL proteins. We determine the structure of the ArhGAP12/G-actin complex, and show that G-actin contacts the RPEL motif and GAP domain sequences. G-actin inhibits ArhGAP12 GAP activity, and this requires the G-actin contacts identified in the structure. In B16 melanoma cells, ArhGAP12 suppresses basal Rac and Cdc42 activity, F-actin assembly, invadopodia formation and experimental metastasis. In this setting, ArhGAP12 mutants defective for G-actin binding exhibit more effective downregulation of Rac GTP loading following HGF stimulation and enhanced inhibition of Rac-dependent processes, including invadopodia formation. Potentiation or disruption of the G-actin/ArhGAP12 interaction, by treatment with the actin-binding drugs latrunculin B or cytochalasin D, has corresponding effects on Rac GTP loading. The interaction of G-actin with RPEL-family rhoGAPs thus provides a negative feedback loop that couples Rac activity to actin dynamics.
Collapse
Affiliation(s)
- Jessica Diring
- Signalling and Transcription Group, The Francis Crick Institute, London, UK
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Neil Q McDonald
- Signalling and Structural Biology Group, The Francis Crick Institute, London, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, UK
| | - Richard Treisman
- Signalling and Transcription Group, The Francis Crick Institute, London, UK.
| |
Collapse
|
12
|
Feng Y, Niu R, Cheng X, Wang K, Du Y, Peng X, Chen F. ATPR-induced differentiation and G0/G1 phase arrest in acute promyelocytic leukemia by repressing EBP50/NCF1 complex to promote the production of ROS. Toxicol Appl Pharmacol 2019; 379:114638. [PMID: 31254567 DOI: 10.1016/j.taap.2019.114638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022]
Abstract
Our previous study has demonstrated that 4-amino-2-trifluoromethyl-phenyl Retinate (ATPR) can induce human leukemia NB4 cells differentiation and G0/G1 phase arrest, but the underlying mechanism is still unclear. In this study, we used proteomics to screen differentially expressed protein profiles in NB4 cells before and after ATPR treatment in vitro. We analyzed the peptides digested from total cellular proteins by reverse phase LC-MS/MS and then performed label-free quantitative analysis. We found 27 significantly up-regulated proteins in the ATPR group compared to the control group. NCF1 was the most significantly changed protein. Immunoprecipitation and double immunofluorescent staining showed that EBP50 bind to NCF1. We further explored the potential molecular mechanism of EBP50/NCF1 complex in ATPR-induced differentiation and G0/G1 phase arrest. The results showed that ATPR remarkably reduced the expression of EBP50 in vivo and in vitro. Interestingly, the reduction of EBP50 contributed to ROS release by modulating the subcellular localization of NCF1. The reduction of EBP50 also contributed to G0/G1 phase arrest by inhibiting CyclinD1, CyclinA2 and CDK4, as well as promoting the differentiation of NB4 cells by increasing the expression of CD11b. Furthermore, we found that the overexpression of EBP50 restrained the effects of ATPR on differentiation and G0/G1 phase arrest in NB4 cells. These results suggest that ATPR-induced differentiation and G0/G1 phase arrest in acute promyelocytic leukemia (APL) by repressing EBP50/NCF1 complex to promote the production of ROS, and the results from in vivo experiments were consistent with those from in vitro studies. Therefore, our finding results suggest that EBP50 may be a new target for ATPR in the treatment of APL.
Collapse
Affiliation(s)
- Yubin Feng
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Ruowen Niu
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Xin Cheng
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Ke Wang
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Yan Du
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Xiaoqing Peng
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Feihu Chen
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
13
|
Saad MH, Rumschlag M, Guerra MH, Savonen CL, Jaster AM, Olson PD, Alazizi A, Luca F, Pique-Regi R, Schmidt CJ, Bannon MJ. Differentially expressed gene networks, biomarkers, long noncoding RNAs, and shared responses with cocaine identified in the midbrains of human opioid abusers. Sci Rep 2019; 9:1534. [PMID: 30733491 PMCID: PMC6367337 DOI: 10.1038/s41598-018-38209-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022] Open
Abstract
Opioid abuse is now the most common cause of accidental death in the US. Although opioids and most other drugs of abuse acutely increase signaling mediated by midbrain dopamine (DA)-synthesizing neurons, little is known about long-lasting changes in DA cells that may contribute to continued opioid abuse, craving, and relapse. A better understanding of the molecular and cellular bases of opioid abuse could lead to advancements in therapeutics. This study comprises, to our knowledge, the first unbiased examination of genome-wide changes in midbrain gene expression associated with human opioid abuse. Our analyses identified differentially expressed genes and distinct gene networks associated with opioid abuse, specific genes with predictive capability for subject assignment to the opioid abuse cohort, and genes most similarly affected in chronic opioid and cocaine abusers. We also identified differentially expressed long noncoding RNAs capable of regulating known drug-responsive protein-coding genes. Opioid-regulated genes identified in this study warrant further investigation as potential biomarkers and/or therapeutic targets for human substance abuse.
Collapse
Affiliation(s)
- Manal H Saad
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA
| | - Matthew Rumschlag
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA
| | - Michael H Guerra
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA
| | - Candace L Savonen
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA
| | - Alaina M Jaster
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA
| | - Philip D Olson
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA
| | - Adnan Alazizi
- Wayne State University School of Medicine, Center for Molecular Medicine & Genetics, Detroit, MI, 48201, USA.,Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, MI, 48201, USA
| | - Francesca Luca
- Wayne State University School of Medicine, Center for Molecular Medicine & Genetics, Detroit, MI, 48201, USA.,Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, MI, 48201, USA
| | - Roger Pique-Regi
- Wayne State University School of Medicine, Center for Molecular Medicine & Genetics, Detroit, MI, 48201, USA.,Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, MI, 48201, USA
| | - Carl J Schmidt
- University of Michigan School of Medicine, Department of Pathology, Detroit, MI, 48109, USA
| | - Michael J Bannon
- Wayne State University School of Medicine, Department of Pharmacology, Detroit, MI, 48201, USA.
| |
Collapse
|
14
|
Rho GTPases in Intellectual Disability: From Genetics to Therapeutic Opportunities. Int J Mol Sci 2018; 19:ijms19061821. [PMID: 29925821 PMCID: PMC6032284 DOI: 10.3390/ijms19061821] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 12/22/2022] Open
Abstract
Rho-class small GTPases are implicated in basic cellular processes at nearly all brain developmental steps, from neurogenesis and migration to axon guidance and synaptic plasticity. GTPases are key signal transducing enzymes that link extracellular cues to the neuronal responses required for the construction of neuronal networks, as well as for synaptic function and plasticity. Rho GTPases are highly regulated by a complex set of activating (GEFs) and inactivating (GAPs) partners, via protein:protein interactions (PPI). Misregulated RhoA, Rac1/Rac3 and cdc42 activity has been linked with intellectual disability (ID) and other neurodevelopmental conditions that comprise ID. All genetic evidences indicate that in these disorders the RhoA pathway is hyperactive while the Rac1 and cdc42 pathways are consistently hypoactive. Adopting cultured neurons for in vitro testing and specific animal models of ID for in vivo examination, the endophenotypes associated with these conditions are emerging and include altered neuronal networking, unbalanced excitation/inhibition and altered synaptic activity and plasticity. As we approach a clearer definition of these phenotype(s) and the role of hyper- and hypo-active GTPases in the construction of neuronal networks, there is an increasing possibility that selective inhibitors and activators might be designed via PPI, or identified by screening, that counteract the misregulation of small GTPases and result in alleviation of the cognitive condition. Here we review all knowledge in support of this possibility.
Collapse
|
15
|
Heun-Johnson H, Levitt P. Differential impact of Met receptor gene interaction with early-life stress on neuronal morphology and behavior in mice. Neurobiol Stress 2017; 8:10-20. [PMID: 29255778 PMCID: PMC5723381 DOI: 10.1016/j.ynstr.2017.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/24/2017] [Accepted: 11/25/2017] [Indexed: 01/01/2023] Open
Abstract
Early adversity in childhood increases the risk of anxiety, mood, and post-traumatic stress disorders in adulthood, and specific gene-by-environment interactions may increase risk further. A common functional variant in the promoter region of the gene encoding the human MET receptor tyrosine kinase (rs1858830 ‘C’ allele) reduces expression of MET and is associated with altered cortical circuit function and structural connectivity. Mice with reduced Met expression exhibit changes in anxiety-like and conditioned fear behavior, precocious synaptic maturation in the hippocampus, and reduced neuronal arbor complexity and synaptogenesis. These phenotypes also can be produced independently by early adversity in wild-type mice. The present study addresses the outcome of combining early-life stress and genetic influences that alter timing of maturation on enduring functional and structural phenotypes. Using a model of reduced Met expression (Met+/−) and early-life stress from postnatal day 2–9, social, anxiety-like, and contextual fear behaviors in later life were measured. Mice that experienced early-life stress exhibited impairments in social interaction, whereas alterations in anxiety-like behavior and fear learning were driven by Met haploinsufficiency, independent of rearing condition. Early-life stress or reduced Met expression decreased arbor complexity of ventral hippocampal CA1 pyramidal neurons projecting to basolateral amygdala. Paradoxically, arbor complexity in Met+/− mice was increased following early-life stress, and thus not different from arbors in wild-type mice raised in control conditions. The changes in dendritic morphology are consistent with the hypothesis that the physiological state of maturation of CA1 neurons in Met+/− mice influences their responsiveness to early-life stress. The dissociation of behavioral and structural changes suggests that there may be phenotype-specific sensitivities to early-life stress.
Collapse
Affiliation(s)
- Hanke Heun-Johnson
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Pat Levitt
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Ba W, Nadif Kasri N. RhoGTPases at the synapse: An embarrassment of choice. Small GTPases 2017; 8:106-113. [PMID: 27492682 PMCID: PMC5464131 DOI: 10.1080/21541248.2016.1206352] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022] Open
Abstract
Activity-dependent modifications in the strength of excitatory synapses are considered to be major cellular mechanisms that contribute to the plasticity of neuronal networks underlying learning and memory. Key mechanisms for the regulation of synaptic efficacy involve the dynamic changes in size and number of dendritic spines, as well as the synaptic incorporation and removal of AMPA-type glutamate receptors (AMPAr). As key regulators of the actin cytoskeleton, the Rho subfamily of GTP-binding proteins play a critical role in synaptic development and plasticity. They shuttle between the active GTP-bound form and the inactive GDP-bound form under the regulation of dedicated guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). More than 80 human GEFs and 70 GAPs have been identified, most of which are expressed in the brain with a specific spatial and temporal expression pattern. However, the function of most GEFs and GAPs in the brain has not been elucidated. In this review, we highlight the novel neuronal function of the synaptic RhoGAP ARHGAP12 and the ID-associated RhoGEF TRIO and further propose 3 possible approaches of neurons utilizing Rho GTPase regulatory proteins to accurately modulate synaptic function.
Collapse
Affiliation(s)
- W. Ba
- Department of Cognitive Neuroscience, Radboudumc, Nijmegen, the Netherlands
- Department of Human Genetics, Radboudumc, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| | - N. Nadif Kasri
- Department of Cognitive Neuroscience, Radboudumc, Nijmegen, the Netherlands
- Department of Human Genetics, Radboudumc, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| |
Collapse
|
17
|
Important roles of Vilse in dendritic architecture and synaptic plasticity. Sci Rep 2017; 7:45646. [PMID: 28368047 PMCID: PMC5377306 DOI: 10.1038/srep45646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/28/2017] [Indexed: 11/17/2022] Open
Abstract
Vilse/Arhgap39 is a Rho GTPase activating protein (RhoGAP) and utilizes its WW domain to regulate Rac/Cdc42-dependent morphogenesis in Drosophila and murine hippocampal neurons. However, the function of Vilse in mammalian dendrite architecture and synaptic plasticity remained unclear. In the present study, we aimed to explore the possible role of Vilse in dendritic structure and synaptic function in the brain. Homozygous knockout of Vilse resulted in premature embryonic lethality in mice. Changes in dendritic complexity and spine density were noticed in hippocampal neurons of Camk2a-Cre mediated forebrain-specific Vilse knockout (VilseΔ/Δ) mice. VilseΔ/Δ mice displayed impaired spatial memory in water maze and Y-maze tests. Electrical stimulation in hippocampal CA1 region revealed that the synaptic transmission and plasticity were defected in VilseΔ/Δ mice. Collectively, our results demonstrate that Vilse is essential for embryonic development and required for spatial memory.
Collapse
|
18
|
Martin-Vilchez S, Whitmore L, Asmussen H, Zareno J, Horwitz R, Newell-Litwa K. RhoGTPase Regulators Orchestrate Distinct Stages of Synaptic Development. PLoS One 2017; 12:e0170464. [PMID: 28114311 PMCID: PMC5256999 DOI: 10.1371/journal.pone.0170464] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 01/05/2017] [Indexed: 11/19/2022] Open
Abstract
Small RhoGTPases regulate changes in post-synaptic spine morphology and density that support learning and memory. They are also major targets of synaptic disorders, including Autism. Here we sought to determine whether upstream RhoGTPase regulators, including GEFs, GAPs, and GDIs, sculpt specific stages of synaptic development. The majority of examined molecules uniquely regulate either early spine precursor formation or later maturation. Specifically, an activator of actin polymerization, the Rac1 GEF β-PIX, drives spine precursor formation, whereas both FRABIN, a Cdc42 GEF, and OLIGOPHRENIN-1, a RhoA GAP, regulate spine precursor elongation. However, in later development, a novel Rac1 GAP, ARHGAP23, and RhoGDIs inactivate actomyosin dynamics to stabilize mature synapses. Our observations demonstrate that specific combinations of RhoGTPase regulatory proteins temporally balance RhoGTPase activity during post-synaptic spine development.
Collapse
Affiliation(s)
- Samuel Martin-Vilchez
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Leanna Whitmore
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Hannelore Asmussen
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Jessica Zareno
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Karen Newell-Litwa
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, United States of America
| |
Collapse
|
19
|
Frega M, van Gestel SHC, Linda K, van der Raadt J, Keller J, Van Rhijn JR, Schubert D, Albers CA, Nadif Kasri N. Rapid Neuronal Differentiation of Induced Pluripotent Stem Cells for Measuring Network Activity on Micro-electrode Arrays. J Vis Exp 2017. [PMID: 28117798 PMCID: PMC5407693 DOI: 10.3791/54900] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neurons derived from human induced Pluripotent Stem Cells (hiPSCs) provide a promising new tool for studying neurological disorders. In the past decade, many protocols for differentiating hiPSCs into neurons have been developed. However, these protocols are often slow with high variability, low reproducibility, and low efficiency. In addition, the neurons obtained with these protocols are often immature and lack adequate functional activity both at the single-cell and network levels unless the neurons are cultured for several months. Partially due to these limitations, the functional properties of hiPSC-derived neuronal networks are still not well characterized. Here, we adapt a recently published protocol that describes production of human neurons from hiPSCs by forced expression of the transcription factor neurogenin-212. This protocol is rapid (yielding mature neurons within 3 weeks) and efficient, with nearly 100% conversion efficiency of transduced cells (>95% of DAPI-positive cells are MAP2 positive). Furthermore, the protocol yields a homogeneous population of excitatory neurons that would allow the investigation of cell-type specific contributions to neurological disorders. We modified the original protocol by generating stably transduced hiPSC cells, giving us explicit control over the total number of neurons. These cells are then used to generate hiPSC-derived neuronal networks on micro-electrode arrays. In this way, the spontaneous electrophysiological activity of hiPSC-derived neuronal networks can be measured and characterized, while retaining interexperimental consistency in terms of cell density. The presented protocol is broadly applicable, especially for mechanistic and pharmacological studies on human neuronal networks.
Collapse
Affiliation(s)
- Monica Frega
- Department of Cognitive Neurosciences, Radboudumc; Donders Institute for Brain, Cognition and Behaviour, Radboud University
| | | | - Katrin Linda
- Donders Institute for Brain, Cognition and Behaviour, Radboud University; Department of Human Genetics, Radboudumc
| | | | - Jason Keller
- Department of Cognitive Neurosciences, Radboudumc; Donders Institute for Brain, Cognition and Behaviour, Radboud University
| | - Jon-Ruben Van Rhijn
- Department of Cognitive Neurosciences, Radboudumc; Donders Institute for Brain, Cognition and Behaviour, Radboud University
| | - Dirk Schubert
- Department of Cognitive Neurosciences, Radboudumc; Donders Institute for Brain, Cognition and Behaviour, Radboud University
| | - Cornelis A Albers
- Donders Institute for Brain, Cognition and Behaviour, Radboud University; Department of Human Genetics, Radboudumc; Department of Molecular Developmental Biology, Radboud University;
| | - Nael Nadif Kasri
- Department of Cognitive Neurosciences, Radboudumc; Donders Institute for Brain, Cognition and Behaviour, Radboud University; Department of Human Genetics, Radboudumc;
| |
Collapse
|