1
|
Sjekloća L, Buratti E. Conserved region of human TDP-43 is structurally similar to membrane binding protein FARP1 and protein chaperons BAG6 and CYP33. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001388. [PMID: 39583578 PMCID: PMC11582883 DOI: 10.17912/micropub.biology.001388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Transactive response DNA-binding protein of 43 KDa (TDP-43) is important for RNA metabolism in all animals and in humans is involved in neuromuscular diseases. Full-length TDP-43 is prone to oligomerization and misfolding what renders difficult its characterization. We report that TDP-43 domains are structurally similar to lipid binding protein FARP1 and protein chaperons BAG6 and CYP33. Sequence analysis suggests putative lipid binding sites throughout TDP-43 and in vitro thioflavin T fluorescence assays show that cholesterol and phosphatidylcholine affect fibrillation of recombinant TDP-43 fragments. Our findings suggest that TDP-43 can bind lipids directly and it may contribute to its own chaperoning.
Collapse
Affiliation(s)
- Ljiljana Sjekloća
- Molecular Pathology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Emanuele Buratti
- Molecular Pathology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
2
|
Domínguez-Berzosa L, Cantarero L, Rodríguez-Sanz M, Tort G, Garrido E, Troya-Balseca J, Sáez M, Castro-Martínez XH, Fernandez-Lizarbe S, Urquizu E, Calvo E, López JA, Palomo T, Palau F, Hoenicka J. ANKK1 Is a Wnt/PCP Scaffold Protein for Neural F-ACTIN Assembly. Int J Mol Sci 2024; 25:10705. [PMID: 39409035 PMCID: PMC11477271 DOI: 10.3390/ijms251910705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
The TaqIA polymorphism is a marker of both the Ankyrin Repeat and Kinase Domain containing I gene (ANKK1) encoding a RIP-kinase, and the DRD2 gene for the dopamine receptor D2. Despite a large number of studies of TaqIA in addictions and other psychiatric disorders, there is difficulty in interpreting this genetic phenomenon due to the lack of knowledge about ANKK1 function. In SH-SY5Y neuroblastoma models, we show that ANKK1 interacts with the synapse protein FERM ARH/RhoGEF and Pleckstrin Domain 1 (FARP1), which is a guanine nucleotide exchange factor (GEF) of the RhoGTPases RAC1 and RhoA. ANKK1-FARP1 colocalized in F-ACTIN-rich structures for neuronal maturation and migration, and both proteins activate the Wnt/PCP pathway. ANKK1, but not FARP1, promotes neuritogenesis, and both proteins are involved in neuritic spine outgrowth. Notably, the knockdown of ANKK1 or FARP1 affects RhoGTPases expression and neural differentiation. Additionally, ANKK1 binds WGEF, another GEF of Wnt/PCP, regulating its interaction with RhoA. During neuronal differentiation, ANKK1-WGEF interaction is downregulated, while ANKK1-FARP1 interaction is increased, suggesting that ANKK1 recruits Wnt/PCP components for bidirectional control of F-ACTIN assembly. Our results suggest a brain structural basis in TaqIA-associated phenotypes.
Collapse
Affiliation(s)
- Laura Domínguez-Berzosa
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 08950 Barcelona, Spain
| | - Lara Cantarero
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 08950 Barcelona, Spain
| | - María Rodríguez-Sanz
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
| | - Gemma Tort
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
| | - Elena Garrido
- Laboratory of Neurosciences, Psychiatry Department, Instituto de Investigación Sanitaria del Hospital Universitario 12 de Octubre, Avda. Andalucía s/n, 28041 Madrid, Spain (T.P.)
| | - Johanna Troya-Balseca
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
| | - María Sáez
- Centro de Investigación Príncipe Felipe (CIPF), 45012 Valencia, Spain; (M.S.); (S.F.-L.)
| | - Xóchitl Helga Castro-Martínez
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
| | - Sara Fernandez-Lizarbe
- Centro de Investigación Príncipe Felipe (CIPF), 45012 Valencia, Spain; (M.S.); (S.F.-L.)
| | - Edurne Urquizu
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
| | - Enrique Calvo
- Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (E.C.); (J.A.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain
| | - Juan Antonio López
- Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (E.C.); (J.A.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain
| | - Tomás Palomo
- Laboratory of Neurosciences, Psychiatry Department, Instituto de Investigación Sanitaria del Hospital Universitario 12 de Octubre, Avda. Andalucía s/n, 28041 Madrid, Spain (T.P.)
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28041 Madrid, Spain
| | - Francesc Palau
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 08950 Barcelona, Spain
- ÚNICAS SJD Center, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Division of Pediatrics, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Janet Hoenicka
- Laboratory of Neurogenetics and Molecular Medicine, Center for Genomic Sciences in Medicine, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (L.D.-B.); (L.C.); (M.R.-S.); (G.T.); (J.T.-B.); (X.H.C.-M.); (E.U.); (F.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 08950 Barcelona, Spain
| |
Collapse
|
3
|
Wiseman S. In conversation with Lucas Cheadle. Nat Neurosci 2024; 27:1428-1431. [PMID: 39107592 DOI: 10.1038/s41593-024-01719-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
|
4
|
Dent LG, Curry N, Sparks H, Bousgouni V, Maioli V, Kumar S, Munro I, Butera F, Jones I, Arias-Garcia M, Rowe-Brown L, Dunsby C, Bakal C. Environmentally dependent and independent control of 3D cell shape. Cell Rep 2024; 43:114016. [PMID: 38636520 DOI: 10.1016/j.celrep.2024.114016] [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: 01/30/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
How cancer cells determine their shape in response to three-dimensional (3D) geometric and mechanical cues is unclear. We develop an approach to quantify the 3D cell shape of over 60,000 melanoma cells in collagen hydrogels using high-throughput stage-scanning oblique plane microscopy (ssOPM). We identify stereotypic and environmentally dependent changes in shape and protrusivity depending on whether a cell is proximal to a flat and rigid surface or is embedded in a soft environment. Environmental sensitivity metrics calculated for small molecules and gene knockdowns identify interactions between the environment and cellular factors that are important for morphogenesis. We show that the Rho guanine nucleotide exchange factor (RhoGEF) TIAM2 contributes to shape determination in environmentally independent ways but that non-muscle myosin II, microtubules, and the RhoGEF FARP1 regulate shape in ways dependent on the microenvironment. Thus, changes in cancer cell shape in response to 3D geometric and mechanical cues are modulated in both an environmentally dependent and independent fashion.
Collapse
Affiliation(s)
- Lucas G Dent
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Nathan Curry
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Hugh Sparks
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Vicky Bousgouni
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Vincent Maioli
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Sunil Kumar
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Ian Munro
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Francesca Butera
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Ian Jones
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Mar Arias-Garcia
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Leo Rowe-Brown
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Chris Dunsby
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK.
| | - Chris Bakal
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
5
|
Rozofsky JP, Pozzuto JM, Byrd-Jacobs CA. Mitral Cell Dendritic Morphology in the Adult Zebrafish Olfactory Bulb following Growth, Injury and Recovery. Int J Mol Sci 2024; 25:5030. [PMID: 38732248 PMCID: PMC11084181 DOI: 10.3390/ijms25095030] [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: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024] Open
Abstract
The role of afferent target interactions in dendritic plasticity within the adult brain remains poorly understood. There is a paucity of data regarding the effects of deafferentation and subsequent dendritic recovery in adult brain structures. Moreover, although adult zebrafish demonstrate ongoing growth, investigations into the impact of growth on mitral cell (MC) dendritic arbor structure and complexity are lacking. Leveraging the regenerative capabilities of the zebrafish olfactory system, we conducted a comprehensive study to address these gaps. Employing an eight-week reversible deafferentation injury model followed by retrograde labeling, we observed substantial morphological alterations in MC dendrites. Our hypothesis posited that cessation of injury would facilitate recovery of MC dendritic arbor structure and complexity, potentially influenced by growth dynamics. Statistical analyses revealed significant changes in MC dendritic morphology following growth and recovery periods, indicating that MC total dendritic branch length retained significance after 8 weeks of deafferentation injury when normalized to individual fish physical characteristics. This suggests that regeneration of branch length could potentially function relatively independently of growth-related changes. These findings underscore the remarkable plasticity of adult dendritic arbor structures in a sophisticated model organism and highlight the efficacy of zebrafish as a vital implement for studying neuroregenerative processes.
Collapse
Affiliation(s)
- John P. Rozofsky
- Department of Biological Sciences, Western Michigan University, 1903 W Michigan Ave., Kalamazoo, MI 49009, USA;
| | - Joanna M. Pozzuto
- Department of Biology, Kalamazoo Valley Community College, 6767 W O Ave., Kalamazoo, MI 49009, USA;
| | - Christine A. Byrd-Jacobs
- Department of Biological Sciences, Western Michigan University, 1903 W Michigan Ave., Kalamazoo, MI 49009, USA;
| |
Collapse
|
6
|
Song Q, Geng H, Zhen H, Liu H, Deng H, Yuan Z, Zhang J, Cao Z, Pang Q, Zhao B. DjFARP Contributes to the Regeneration and Maintenance of the Brain through Activation of DjRac1 in Dugesia japonica. Mol Neurobiol 2023; 60:6294-6306. [PMID: 37442859 DOI: 10.1007/s12035-023-03478-6] [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: 10/09/2022] [Accepted: 07/02/2023] [Indexed: 07/15/2023]
Abstract
FERM, RhoGEF, and Pleckstrin domain protein (FARP) mediated RhoGTPase pathways are involved in diverse biological processes, such as neuronal development and tumorigenesis. However, little is known about their role in neural regeneration. We uncovered for the first time that FARP-Rac1 signaling plays an important role in neural regeneration in Dugesia japonica, a planarian that possesses unparalleled regenerative capacities. The planarian FARP homolog DjFARP was primarily expressed in both intact and regenerating brain and pharynx tissue. Functional studies suggested that downregulation of DjFARP with dsRNA in Dugesia japonica led to smaller brain sizes, defects in brain lateral branches, and loss of cholinergic, GABAergic, and dopaminergic neurons in both intact and regenerating animals. Moreover, the Rho GTPase DjRac1 was shown to play a similar role in neural regeneration and maintenance. Rac1 activation assay showed that DjFARP acts as a guanine nucleotide exchange factor (GEF) for DjRac1. Together, these findings indicate that the brain defects seen in DjFARP knockdown animals may be attributable to DjRac1 inactivation. In conclusion, our study demonstrated that DjFARP-DjRac1 signaling was required for the maintenance and proper regeneration of the brain in Dugesia japonica.
Collapse
Affiliation(s)
- Qian Song
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, 266 Xincun West Road, Zibo, 255049, People's Republic of China
| | - Huazhi Geng
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Hui Zhen
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, 266 Xincun West Road, Zibo, 255049, People's Republic of China
| | - Hongjin Liu
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Hongkuan Deng
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Zuoqing Yuan
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Jianyong Zhang
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Zhonghong Cao
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Qiuxiang Pang
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, 266 Xincun West Road, Zibo, 255049, People's Republic of China.
- Zibo Maternal and Child Health Hospital, Zibo, 255000, China.
| |
Collapse
|
7
|
Hua Z, Zhang J, Cheng W, Wang C, Zhao D. Ethanolic Extract from Seed Residues of Sea Buckthorn ( Hippophae rhamnoides L.) Ameliorates Oxidative Stress Damage and Prevents Apoptosis in Murine Cell and Aging Animal Models. Foods 2023; 12:3322. [PMID: 37685254 PMCID: PMC10487224 DOI: 10.3390/foods12173322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/21/2023] [Accepted: 09/02/2023] [Indexed: 09/10/2023] Open
Abstract
Hippophae rhamnoides L. has been widely used in research and application for almost two decades. While significant progress was achieved in the examination of its fruits and seeds, the exploration and utilization of its by-products have received relatively less attention. This study aims to address this research gap by investigating the effects and underlying mechanisms of sea buckthorn seed residues both in vitro and in vivo. The primary objective of this study is to assess the potential of the hydroalcoholic extract from sea buckthorn seed residues (HYD-SBSR) to prevent cell apoptosis and mitigate oxidative stress damage. To achieve this, an H2O2-induced B16F10 cell model and a D-galactose-induced mouse model were used. The H2O2-induced oxidative stress model using B16F10 cells was utilized to evaluate the cellular protective and reparative effects of HYD-SBSR. The results demonstrated the cytoprotective effects of HYD-SBSR, as evidenced by reduced apoptosis rates and enhanced resistance to oxidative stress alongside moderate cell repair properties. Furthermore, this study investigated the impact of HYD-SBSR on antioxidant enzymes and peroxides in mice to elucidate its reparative potential in vivo. The findings revealed that HYD-SBSR exhibited remarkable antioxidant performance, particularly at low concentrations, significantly enhancing antioxidant capacity under oxidative stress conditions. To delve into the mechanisms underlying HYD-SBSR, a comprehensive proteomics analysis was conducted to identify differentially expressed proteins (DEPs). Additionally, a Gene Ontology (GO) analysis and an Encyclopedia of Genes and Genomes (KEGG) pathway cluster analysis were performed to elucidate the functional roles of these DEPs. The outcomes highlighted crucial mechanistic pathways associated with HYD-SBSR, including the PPAR signaling pathway, fat digestion and absorption, glycerophospholipid metabolism, and cholesterol metabolism. The research findings indicated that HYD-SBSR, as a health food supplement, exhibits favorable effects by promoting healthy lipid metabolism, contributing to the sustainable and environmentally friendly production of sea buckthorn and paving the way for future investigations and applications in the field of nutraceutical and pharmaceutical research.
Collapse
Affiliation(s)
- Zhongjie Hua
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing 100048, China
| | - Jiachan Zhang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing 100048, China
| | - Wenjing Cheng
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing 100048, China
| | - Changtao Wang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing 100048, China
| | - Dan Zhao
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
8
|
Cucinotta F, Lintas C, Tomaiuolo P, Baccarin M, Picinelli C, Castronovo P, Sacco R, Piras IS, Turriziani L, Ricciardello A, Scattoni ML, Persico AM. Diagnostic yield and clinical impact of chromosomal microarray analysis in autism spectrum disorder. Mol Genet Genomic Med 2023; 11:e2182. [PMID: 37186221 PMCID: PMC10422062 DOI: 10.1002/mgg3.2182] [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: 10/10/2022] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is characterized by high heritability estimates and recurrence rates; its genetic underpinnings are very heterogeneous and include variable combinations of common and rare variants. Array-comparative genomic hybridization (aCGH) offers significant sensitivity for the identification of copy number variants (CNVs), which can act as susceptibility or causal factors for ASD. METHODS The aim of this study was to evaluate both diagnostic yield and clinical impact of aCGH in 329 ASD patients of Italian descent. RESULTS Pathogenic/likely pathogenic CNVs were identified in 50/329 (15.2%) patients, whereas 89/329 (27.1%) carry variants of uncertain significance. The 10 most enriched gene sets identified by Gene Ontology Enrichment Analysis are primarily involved in neuronal function and synaptic connectivity. In 13/50 (26.0%) patients with pathogenic/likely pathogenic CNVs, the outcome of array-CGH led to the request of 25 additional medical exams which would not have otherwise been prescribed, mainly including brain MRI, EEG, EKG, and/or cardiac ultrasound. A positive outcome was obtained in 12/25 (48.0%) of these additional tests. CONCLUSIONS This study confirms the satisfactory diagnostic yield of aCGH, underscoring its potential for better, more in-depth care of children with autism when genetic results are analyzed also with a focus on patient management.
Collapse
Affiliation(s)
- Francesca Cucinotta
- Interdepartmental Program "Autism 0‐90", "G. Martino" University Hospital of MessinaMessinaItaly
- IRCCS Centro Neurolesi “Bonino Pulejo”MessinaItaly
| | - Carla Lintas
- Service for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and NeurogeneticsUniversity “Campus Bio‐Medico”RomeItaly
| | - Pasquale Tomaiuolo
- Interdepartmental Program "Autism 0‐90", "G. Martino" University Hospital of MessinaMessinaItaly
| | - Marco Baccarin
- Mafalda Luce Center for Pervasive Developmental DisordersMilanItaly
- Synlab GeneticsBioggioSwitzerland
| | - Chiara Picinelli
- Mafalda Luce Center for Pervasive Developmental DisordersMilanItaly
| | - Paola Castronovo
- Mafalda Luce Center for Pervasive Developmental DisordersMilanItaly
| | - Roberto Sacco
- Service for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and NeurogeneticsUniversity “Campus Bio‐Medico”RomeItaly
| | - Ignazio Stefano Piras
- Service for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and NeurogeneticsUniversity “Campus Bio‐Medico”RomeItaly
- Neurogenomics DivisionThe Translational Genomics Research InstitutePhoenixArizonaUSA
| | - Laura Turriziani
- Interdepartmental Program "Autism 0‐90", "G. Martino" University Hospital of MessinaMessinaItaly
| | - Arianna Ricciardello
- Interdepartmental Program "Autism 0‐90", "G. Martino" University Hospital of MessinaMessinaItaly
| | | | - Antonio M. Persico
- Child and Adolescent Neuropsychiatry Program, Modena University Hospital & Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| |
Collapse
|
9
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538959. [PMID: 37205520 PMCID: PMC10187231 DOI: 10.1101/2023.05.01.538959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNAseq data ob-tained from E12.5 fetal mouse brains 3 hours after VPA administration revealed that VPA significant-ly increased or decreased the expression of approximately 7,300 genes. No significant sex differ-ences in VPA-induced gene expression were observed. Expression of genes associated with neu-rodevelopmental disorders (NDDs) such as autism as well as neurogenesis, axon growth and syn-aptogenesis, GABAergic, glutaminergic and dopaminergic synaptic transmission, perineuronal nets, and circadian rhythms was dysregulated by VPA. Moreover, expression of 399 autism risk genes was significantly altered by VPA as was expression of 252 genes that have been reported to play fundamental roles in the development of the nervous system but are not otherwise linked to autism. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity in the postnatal and adult brain. The set of genes meeting these criteria pro-vides potential targets for future hypothesis-driven approaches to elucidating the proximal underly-ing causes of defective brain connectivity in NDDs such as autism.
Collapse
|
10
|
Mizumoto K, Jin Y, Bessereau JL. Synaptogenesis: unmasking molecular mechanisms using Caenorhabditis elegans. Genetics 2023; 223:iyac176. [PMID: 36630525 PMCID: PMC9910414 DOI: 10.1093/genetics/iyac176] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/22/2022] [Indexed: 01/13/2023] Open
Abstract
The nematode Caenorhabditis elegans is a research model organism particularly suited to the mechanistic understanding of synapse genesis in the nervous system. Armed with powerful genetics, knowledge of complete connectomics, and modern genomics, studies using C. elegans have unveiled multiple key regulators in the formation of a functional synapse. Importantly, many signaling networks display remarkable conservation throughout animals, underscoring the contributions of C. elegans research to advance the understanding of our brain. In this chapter, we will review up-to-date information of the contribution of C. elegans to the understanding of chemical synapses, from structure to molecules and to synaptic remodeling.
Collapse
Affiliation(s)
- Kota Mizumoto
- Department of Zoology, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Yishi Jin
- Department of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Jean-Louis Bessereau
- Univ Lyon, University Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U 1314, Melis, 69008 Lyon, France
| |
Collapse
|
11
|
Jahan MS, Tsuzuki T, Ito T, Bhuiyan MER, Takahashi I, Takamatsu H, Kumanogoh A, Negishi T, Yukawa K. PlexinA1-deficient mice exhibit decreased cell density and augmented oxidative stress in parvalbumin-expressing interneurons in the medial prefrontal cortex. IBRO Neurosci Rep 2022; 13:500-512. [DOI: 10.1016/j.ibneur.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/20/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022] Open
|
12
|
Kazanietz MG, Cooke M, Garcia-Mata R. Nonredundant Rac-GEF control of actin cytoskeleton reorganization. Trends Cell Biol 2022; 32:815-818. [PMID: 35753960 PMCID: PMC9930409 DOI: 10.1016/j.tcb.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 02/08/2023]
Abstract
Rac-GEFs operate in a nonredundant manner as downstream effectors of receptor tyrosine kinases to promote ruffle formation, indicative of unique modes of regulation and targeting. Current research is shedding light on the intricate signaling paradigms shaping spatiotemporal activation of the small GTPase Rac during the generation of actin-rich membrane protrusions.
Collapse
Affiliation(s)
- Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Einstein Medical Center Philadelphia, Philadelphia, PA 19141, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA.
| |
Collapse
|
13
|
FARP1, ARHGEF39, and TIAM2 are essential receptor tyrosine kinase effectors for Rac1-dependent cell motility in human lung adenocarcinoma. Cell Rep 2021; 37:109905. [PMID: 34731623 PMCID: PMC8627373 DOI: 10.1016/j.celrep.2021.109905] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/27/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the undisputable role of the small GTPase Rac1 in the regulation of actin cytoskeleton reorganization, the Rac guanine-nucleotide exchange factors (Rac-GEFs) involved in Rac1-mediated motility and invasion in human lung adenocarcinoma cells remain largely unknown. Here, we identify FARP1, ARHGEF39, and TIAM2 as essential Rac-GEFs responsible for Rac1-mediated lung cancer cell migration upon EGFR and c-Met activation. Noteworthily, these Rac-GEFs operate in a non-redundant manner by controlling distinctive aspects of ruffle dynamics formation. Mechanistic analysis reveals a leading role of the AXL-Gab1-PI3K axis in conferring pro-motility traits downstream of EGFR. Along with the positive association between the overexpression of Rac-GEFs and poor lung adenocarcinoma patient survival, we show that FARP1 and ARHGEF39 are upregulated in EpCam+ cells sorted from primary human lung adenocarcinomas. Overall, our study reveals fundamental insights into the complex intricacies underlying Rac-GEF-mediated cancer cell motility signaling, hence underscoring promising targets for metastatic lung cancer therapy.
Collapse
|
14
|
Modelling and Refining Neuronal Circuits with Guidance Cues: Involvement of Semaphorins. Int J Mol Sci 2021; 22:ijms22116111. [PMID: 34204060 PMCID: PMC8201269 DOI: 10.3390/ijms22116111] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022] Open
Abstract
The establishment of neuronal circuits requires neurons to develop and maintain appropriate connections with cellular partners in and out the central nervous system. These phenomena include elaboration of dendritic arborization and formation of synaptic contacts, initially made in excess. Subsequently, refinement occurs, and pruning takes places both at axonal and synaptic level, defining a homeostatic balance maintained throughout the lifespan. All these events require genetic regulations which happens cell-autonomously and are strongly influenced by environmental factors. This review aims to discuss the involvement of guidance cues from the Semaphorin family.
Collapse
|
15
|
Carulli D, de Winter F, Verhaagen J. Semaphorins in Adult Nervous System Plasticity and Disease. Front Synaptic Neurosci 2021; 13:672891. [PMID: 34045951 PMCID: PMC8148045 DOI: 10.3389/fnsyn.2021.672891] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Semaphorins, originally discovered as guidance cues for developing axons, are involved in many processes that shape the nervous system during development, from neuronal proliferation and migration to neuritogenesis and synapse formation. Interestingly, the expression of many Semaphorins persists after development. For instance, Semaphorin 3A is a component of perineuronal nets, the extracellular matrix structures enwrapping certain types of neurons in the adult CNS, which contribute to the closure of the critical period for plasticity. Semaphorin 3G and 4C play a crucial role in the control of adult hippocampal connectivity and memory processes, and Semaphorin 5A and 7A regulate adult neurogenesis. This evidence points to a role of Semaphorins in the regulation of adult neuronal plasticity. In this review, we address the distribution of Semaphorins in the adult nervous system and we discuss their function in physiological and pathological processes.
Collapse
Affiliation(s)
- Daniela Carulli
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
- Department of Neuroscience Rita Levi-Montalcini and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Turin, Italy
| | - Fred de Winter
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| |
Collapse
|
16
|
Xiaozhen S, Fan Y, Fang Y, Xiaoping L, Jia J, Wuhen X, Xiaojun T, Jun S, Yucai C, Hong Z, Guang H, Shengnan W. Novel Truncating and Missense Variants in SEMA6B in Patients With Early-Onset Epilepsy. Front Cell Dev Biol 2021; 9:633819. [PMID: 34017830 PMCID: PMC8129541 DOI: 10.3389/fcell.2021.633819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/17/2021] [Indexed: 11/13/2022] Open
Abstract
Progressive myoclonic epilepsy (PME) is a rare neurodegenerative disease, characterized by myoclonic seizures and tonic clonic seizures, with genetical and phenotypical heterogeneity. The semaphorin 6B (SEMA6B) gene has been recently reported a causal gene of PME. Independent studies are warranted to further support these findings. Here we report that one nonsense variant in NM_032108.3 exon17 c.2056C > T (p.Gln686∗) and one missense variant in exon14 c.1483G > T (p.Gly495Trp) of SEMA6B, both occurring de novo, underlie early-onset epilepsy with variable severity and different response to treatment in two patients. In vitro analyses have demonstrated that the nonsense variant, p.Gln686∗, results in a truncated protein with remarkably increased expression compared to that of the wild type. The truncated protein presented more homogeneous and failed to locate in the plasma membrane. The missense variant p.Gly495Trp affects evolutionarily conserved amino acid and is located in the sema domain, a key functional domain of SEMA6B. It was predicted to perturb the SEMA6B function by altering the tertiary structure of mutant protein, although neither change of protein length and expression nor difference of cellular distribution was observed. Co-immunoprecipitation studies have demonstrated that both variants influence protein binding of SEMA6B and PlxnA2 with varying degrees. Our results provide further evidence to support the initial findings of SEMA6B being causal to epilepsy and indicate that mediating Semaphorin/Plexin signaling is the potential mechanism of the SEMA6B-related disease.
Collapse
Affiliation(s)
- Song Xiaozhen
- Laboratory of Molecular Diagnosis, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Fan
- Bio-X Institute, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Institute of Mental Health, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Fang
- Department of Neurology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lan Xiaoping
- Laboratory of Molecular Diagnosis, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Jia
- Fuxiang Gene Engineering Research Institute, Shanghai, China
| | - Xu Wuhen
- Laboratory of Molecular Diagnosis, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tang Xiaojun
- Laboratory of Molecular Diagnosis, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shen Jun
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Chen Yucai
- Department of Neurology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhang Hong
- Laboratory of Molecular Diagnosis, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - He Guang
- Bio-X Institute, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Institute of Mental Health, Shanghai Jiao Tong University, Shanghai, China
| | - Wu Shengnan
- Laboratory of Molecular Diagnosis, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Lin TY, Chen PJ, Yu HH, Hsu CP, Lee CH. Extrinsic Factors Regulating Dendritic Patterning. Front Cell Neurosci 2021; 14:622808. [PMID: 33519386 PMCID: PMC7838386 DOI: 10.3389/fncel.2020.622808] [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: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Stereotypic dendrite arborizations are key morphological features of neuronal identity, as the size, shape and location of dendritic trees determine the synaptic input fields and how information is integrated within developed neural circuits. In this review, we focus on the actions of extrinsic intercellular communication factors and their effects on intrinsic developmental processes that lead to dendrite patterning. Surrounding neurons or supporting cells express adhesion receptors and secreted proteins that respectively, act via direct contact or over short distances to shape, size, and localize dendrites during specific developmental stages. The different ligand-receptor interactions and downstream signaling events appear to direct dendrite morphogenesis by converging on two categorical mechanisms: local cytoskeletal and adhesion modulation and global transcriptional regulation of key dendritic growth components, such as lipid synthesis enzymes. Recent work has begun to uncover how the coordinated signaling of multiple extrinsic factors promotes complexity in dendritic trees and ensures robust dendritic patterning.
Collapse
Affiliation(s)
- Tzu-Yang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Pei-Ju Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Hsiang Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chi-Hon Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
18
|
Iyer H, Wahul AB, P K A, Sawant BS, Kumar A. A BRD's (BiRD's) eye view of BET and BRPF bromodomains in neurological diseases. Rev Neurosci 2021; 32:403-426. [PMID: 33661583 DOI: 10.1515/revneuro-2020-0067] [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: 07/08/2020] [Accepted: 10/11/2020] [Indexed: 01/18/2023]
Abstract
Neurological disorders (NLDs) are among the top leading causes for disability worldwide. Dramatic changes in the epigenetic topography of the brain and nervous system have been found in many NLDs. Histone lysine acetylation has prevailed as one of the well characterised epigenetic modifications in these diseases. Two instrumental components of the acetylation machinery are the evolutionarily conserved Bromodomain and PHD finger containing (BRPF) and Bromo and Extra terminal domain (BET) family of proteins, also referred to as acetylation 'readers'. Several reasons, including their distinct mechanisms of modulation of gene expression and their property of being highly tractable small molecule targets, have increased their translational relevance. Thus, compounds which demonstrated promising results in targeting these proteins have advanced to clinical trials. They have been established as key role players in pathologies of cancer, cardiac diseases, renal diseases and rheumatic diseases. In addition, studies implicating the role of these bromodomains in NLDs are gaining pace. In this review, we highlight the findings of these studies, and reason for the plausible roles of all BET and BRPF members in NLDs. A comprehensive understanding of their multifaceted functions would be radical in the development of therapeutic interventions.
Collapse
Affiliation(s)
- Harish Iyer
- Epigenetics and Neuropsychiatric Disorders' Laboratory, CSIR - Centre for Cellular and Molecular Biology (CCMB), Hyderabad500007, India
| | - Abhipradnya B Wahul
- Epigenetics and Neuropsychiatric Disorders' Laboratory, CSIR - Centre for Cellular and Molecular Biology (CCMB), Hyderabad500007, India
| | - Annapoorna P K
- Epigenetics and Neuropsychiatric Disorders' Laboratory, CSIR - Centre for Cellular and Molecular Biology (CCMB), Hyderabad500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad201002, India
| | - Bharvi S Sawant
- Epigenetics and Neuropsychiatric Disorders' Laboratory, CSIR - Centre for Cellular and Molecular Biology (CCMB), Hyderabad500007, India
| | - Arvind Kumar
- Epigenetics and Neuropsychiatric Disorders' Laboratory, CSIR - Centre for Cellular and Molecular Biology (CCMB), Hyderabad500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad201002, India
| |
Collapse
|
19
|
Imbalanced post- and extrasynaptic SHANK2A functions during development affect social behavior in SHANK2-mediated neuropsychiatric disorders. Mol Psychiatry 2021; 26:6482-6504. [PMID: 34021263 PMCID: PMC8760046 DOI: 10.1038/s41380-021-01140-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Mutations in SHANK genes play an undisputed role in neuropsychiatric disorders. Until now, research has focused on the postsynaptic function of SHANKs, and prominent postsynaptic alterations in glutamatergic signal transmission have been reported in Shank KO mouse models. Recent studies have also suggested a possible presynaptic function of SHANK proteins, but these remain poorly defined. In this study, we examined how SHANK2 can mediate electrophysiological, molecular, and behavioral effects by conditionally overexpressing either wild-type SHANK2A or the extrasynaptic SHANK2A(R462X) variant. SHANK2A overexpression affected pre- and postsynaptic targets and revealed a reversible, development-dependent autism spectrum disorder-like behavior. SHANK2A also mediated redistribution of Ca2+-permeable AMPA receptors between apical and basal hippocampal CA1 dendrites, leading to impaired synaptic plasticity in the basal dendrites. Moreover, SHANK2A overexpression reduced social interaction and increased the excitatory noise in the olfactory cortex during odor processing. In contrast, overexpression of the extrasynaptic SHANK2A(R462X) variant did not impair hippocampal synaptic plasticity, but still altered the expression of presynaptic/axonal signaling proteins. We also observed an attention-deficit/hyperactivity-like behavior and improved social interaction along with enhanced signal-to-noise ratio in cortical odor processing. Our results suggest that the disruption of pre- and postsynaptic SHANK2 functions caused by SHANK2 mutations has a strong impact on social behavior. These findings indicate that pre- and postsynaptic SHANK2 actions cooperate for normal neuronal function, and that an imbalance between these functions may lead to different neuropsychiatric disorders.
Collapse
|
20
|
Ward J, Tunbridge EM, Sandor C, Lyall LM, Ferguson A, Strawbridge RJ, Lyall DM, Cullen B, Graham N, Johnston KJA, Webber C, Escott-Price V, O'Donovan M, Pell JP, Bailey MES, Harrison PJ, Smith DJ. The genomic basis of mood instability: identification of 46 loci in 363,705 UK Biobank participants, genetic correlation with psychiatric disorders, and association with gene expression and function. Mol Psychiatry 2020; 25:3091-3099. [PMID: 31168069 PMCID: PMC7116257 DOI: 10.1038/s41380-019-0439-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/03/2019] [Accepted: 04/29/2019] [Indexed: 01/01/2023]
Abstract
Genome-wide association studies (GWAS) of psychiatric phenotypes have tended to focus on categorical diagnoses, but to understand the biology of mental illness it may be more useful to study traits which cut across traditional boundaries. Here, we report the results of a GWAS of mood instability as a trait in a large population cohort (UK Biobank, n = 363,705). We also assess the clinical and biological relevance of the findings, including whether genetic associations show enrichment for nervous system pathways. Forty six unique loci associated with mood instability were identified with a SNP heritability estimate of 9%. Linkage Disequilibrium Score Regression (LDSR) analyses identified genetic correlations with Major Depressive Disorder (MDD), Bipolar Disorder (BD), Schizophrenia, anxiety, and Post Traumatic Stress Disorder (PTSD). Gene-level and gene set analyses identified 244 significant genes and 6 enriched gene sets. Tissue expression analysis of the SNP-level data found enrichment in multiple brain regions, and eQTL analyses highlighted an inversion on chromosome 17 plus two brain-specific eQTLs. In addition, we used a Phenotype Linkage Network (PLN) analysis and community analysis to assess for enrichment of nervous system gene sets using mouse orthologue databases. The PLN analysis found enrichment in nervous system PLNs for a community containing serotonin and melatonin receptors. In summary, this work has identified novel loci, tissues and gene sets contributing to mood instability. These findings may be relevant for the identification of novel trans-diagnostic drug targets and could help to inform future stratified medicine innovations in mental health.
Collapse
Affiliation(s)
- Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Cynthia Sandor
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Nicholas Graham
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Caleb Webber
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
- Department of Physiology, Anatomy and Genetics, Oxford, UK
| | | | | | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| |
Collapse
|
21
|
Ouyang L, Chen Y, Wang Y, Chen Y, Fu AKY, Fu WY, Ip NY. p39-associated Cdk5 activity regulates dendritic morphogenesis. Sci Rep 2020; 10:18746. [PMID: 33127972 PMCID: PMC7603351 DOI: 10.1038/s41598-020-75264-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Dendrites, branched structures extending from neuronal cell soma, are specialized for processing information from other neurons. The morphogenesis of dendritic structures is spatiotemporally regulated by well-orchestrated signaling cascades. Dysregulation of these processes impacts the wiring of neuronal circuit and efficacy of neurotransmission, which contribute to the pathogeneses of neurological disorders. While Cdk5 (cyclin-dependent kinase 5) plays a critical role in neuronal dendritic development, its underlying molecular control is not fully understood. In this study, we show that p39, one of the two neuronal Cdk5 activators, is a key regulator of dendritic morphogenesis. Pyramidal neurons deficient in p39 exhibit aberrant dendritic morphology characterized by shorter length and reduced arborization, which is comparable to dendrites in Cdk5-deficient neurons. RNA sequencing analysis shows that the adaptor protein, WDFY1 (WD repeat and FYVE domain-containing 1), acts downstream of Cdk5/p39 to regulate dendritic morphogenesis. While WDFY1 is elevated in p39-deficient neurons, suppressing its expression rescues the impaired dendritic arborization. Further phosphoproteomic analysis suggests that Cdk5/p39 mediates dendritic morphogenesis by modulating various downstream signaling pathways, including PI3K/Akt-, cAMP-, or small GTPase-mediated signaling transduction pathways, thereby regulating cytoskeletal organization, protein synthesis, and protein trafficking.
Collapse
Affiliation(s)
- Li Ouyang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Ye Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Yuewen Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Wing-Yu Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China. .,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China. .,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
22
|
Cucinotta F, Ricciardello A, Turriziani L, Calabrese G, Briguglio M, Boncoddo M, Bellomo F, Tomaiuolo P, Martines S, Bruschetta M, La Fauci Belponer F, Di Bella T, Colombi C, Baccarin M, Picinelli C, Castronovo P, Lintas C, Sacco R, Biederer T, Kellam B, Scherer SW, Persico AM. FARP-1 deletion is associated with lack of response to autism treatment by early start denver model in a multiplex family. Mol Genet Genomic Med 2020; 8:e1373. [PMID: 32588496 PMCID: PMC7507005 DOI: 10.1002/mgg3.1373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/28/2020] [Accepted: 05/31/2020] [Indexed: 01/05/2023] Open
Abstract
Background Children with autism spectrum disorder (ASD) display impressive clinical heterogeneity, also involving treatment response. Genetic variants can contribute to explain this large interindividual phenotypic variability. Methods Array‐CGH (a‐CGH) and whole genome sequencing (WGS) were performed on a multiplex family with two small children diagnosed with ASD at 17 and 18 months of age. Both brothers received the same naturalistic intervention for one year according to the Early Start Denver Model (ESDM), applied by the same therapists, yielding dramatically different treatment outcomes. Results The older sibling came out of the autism spectrum, while the younger sibling displayed very little, in any, improvement. This boy was subsequently treated applying a structured Early Intensive Behavioral Intervention paired with Augmentative Alternative Communication, which yielded a partial response within another year. The ESDM nonresponsive child carries a novel maternally inherited 65 Kb deletion at chr. 13q32.2 spanning FARP1. Farp1 is a synaptic scaffolding protein, which plays a significant role in neural plasticity. Conclusion These results represent a paradigmatic example of the heuristic potential of genetic markers in predicting treatment response and possibly in supporting the targeted prescription of specific early intervention approaches.
Collapse
Affiliation(s)
- Francesca Cucinotta
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Arianna Ricciardello
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Laura Turriziani
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Giorgia Calabrese
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Marilena Briguglio
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Maria Boncoddo
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Fabiana Bellomo
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Pasquale Tomaiuolo
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Silvia Martines
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Marianna Bruschetta
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | | | - Tiziana Di Bella
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| | - Costanza Colombi
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Marco Baccarin
- Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | - Chiara Picinelli
- Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | - Paola Castronovo
- Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | - Carla Lintas
- Service for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy
| | - Roberto Sacco
- Service for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Rome, Italy
| | - Thomas Biederer
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Barbara Kellam
- Genetics & Genome Biology Program, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Stephen W Scherer
- Genetics & Genome Biology Program, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,McLaughlin Centre, University of Toronto, Toronto, Canada
| | - Antonio M Persico
- Interdepartmental Program "Autism 0-90", "G. Martino" University Hospital of Messina, Messina, Italy
| |
Collapse
|
23
|
Modular and Distinct Plexin-A4/FARP2/Rac1 Signaling Controls Dendrite Morphogenesis. J Neurosci 2020; 40:5413-5430. [PMID: 32499377 DOI: 10.1523/jneurosci.2730-19.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/29/2020] [Accepted: 05/26/2020] [Indexed: 12/26/2022] Open
Abstract
Diverse neuronal populations with distinct cellular morphologies coordinate the complex function of the nervous system. Establishment of distinct neuronal morphologies critically depends on signaling pathways that control axonal and dendritic development. The Sema3A-Nrp1/PlxnA4 signaling pathway promotes cortical neuron basal dendrite arborization but also repels axons. However, the downstream signaling components underlying these disparate functions of Sema3A signaling are unclear. Using the novel PlxnA4KRK-AAA knock-in male and female mice, generated by CRISPR/cas9, we show here that the KRK motif in the PlxnA4 cytoplasmic domain is required for Sema3A-mediated cortical neuron dendritic elaboration but is dispensable for inhibitory axon guidance. The RhoGEF FARP2, which binds to the KRK motif, shows identical functional specificity as the KRK motif in the PlxnA4 receptor. We find that Sema3A activates the small GTPase Rac1, and that Rac1 activity is required for dendrite elaboration but not axon growth cone collapse. This work identifies a novel Sema3A-Nrp1/PlxnA4/FARP2/Rac1 signaling pathway that specifically controls dendritic morphogenesis but is dispensable for repulsive guidance events. Overall, our results demonstrate that the divergent signaling output from multifunctional receptor complexes critically depends on distinct signaling motifs, highlighting the modular nature of guidance cue receptors and its potential to regulate diverse cellular responses.SIGNIFICANCE STATEMENT The proper formation of axonal and dendritic morphologies is crucial for the precise wiring of the nervous system that ultimately leads to the generation of complex functions in an organism. The Semaphorin3A-Neuropilin1/Plexin-A4 signaling pathway has been shown to have multiple key roles in neurodevelopment, from axon repulsion to dendrite elaboration. This study demonstrates that three specific amino acids, the KRK motif within the Plexin-A4 receptor cytoplasmic domain, are required to coordinate the downstream signaling molecules to promote Sema3A-mediated cortical neuron dendritic elaboration, but not inhibitory axon guidance. Our results unravel a novel Semaphorin3A-Plexin-A4 downstream signaling pathway and shed light on how the disparate functions of axon guidance and dendritic morphogenesis are accomplished by the same extracellular ligand in vivo.
Collapse
|
24
|
The netrin receptor UNC-40/DCC assembles a postsynaptic scaffold and sets the synaptic content of GABA A receptors. Nat Commun 2020; 11:2674. [PMID: 32471987 PMCID: PMC7260190 DOI: 10.1038/s41467-020-16473-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 04/28/2020] [Indexed: 01/11/2023] Open
Abstract
Increasing evidence indicates that guidance molecules used during development for cellular and axonal navigation also play roles in synapse maturation and homeostasis. In C. elegans the netrin receptor UNC-40/DCC controls the growth of dendritic-like muscle cell extensions towards motoneurons and is required to recruit type A GABA receptors (GABAARs) at inhibitory neuromuscular junctions. Here we show that activation of UNC-40 assembles an intracellular synaptic scaffold by physically interacting with FRM-3, a FERM protein orthologous to FARP1/2. FRM-3 then recruits LIN-2, the ortholog of CASK, that binds the synaptic adhesion molecule NLG-1/Neuroligin and physically connects GABAARs to prepositioned NLG-1 clusters. These processes are orchestrated by the synaptic organizer CePunctin/MADD-4, which controls the localization of GABAARs by positioning NLG-1/neuroligin at synapses and regulates the synaptic content of GABAARs through the UNC-40-dependent intracellular scaffold. Since DCC is detected at GABA synapses in mammals, DCC might also tune inhibitory neurotransmission in the mammalian brain. The netrin receptor UNC-40/DCC is required to recruit GABAAR at neuromuscular junctions in C. elegans. Here, the authors show that UNC-40/DCC assembles an intracellular synaptic scaffold, regulating the content of GABAAR and inhibitory neurotransmission.
Collapse
|
25
|
Semaphorin-Mediated Corticospinal Axon Elimination Depends on the Activity-Induced Bax/Bak-Caspase Pathway. J Neurosci 2020; 40:5402-5412. [PMID: 32471877 DOI: 10.1523/jneurosci.3190-18.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Axon guidance molecules and neuronal activity have been implicated in the establishment and refinement of neural circuits during development. It is unclear, however, whether these guidance molecule- and activity-dependent mechanisms interact with one another to shape neural circuit formation. The formation of corticospinal (CS) circuits, which are essential for voluntary movements, involves both guidance molecule- and activity-dependent components during development. We previously showed that semaphorin6D (Sema6D)-plexinA1 (PlexA1) signaling eliminates ipsilateral projections of CS neurons in the spinal cord, while other studies demonstrate that CS projections to the spinal cord are eliminated in an activity-dependent manner. Here we show that inhibition of cortical neurons during postnatal development causes defects in elimination of ipsilateral CS projections in mice. We further show that mice that lack the activity-dependent Bax/Bak pathway or caspase-9 similarly exhibit defects in elimination of ipsilateral CS projections, suggesting that the activity-dependent Bax/Bak-caspase-9 pathway is essential for the removal of ipsilateral CS projections. Interestingly, either inhibition of neuronal activity in the cortex or deletion of Bax/Bak in mice causes a reduction in PlexA1 protein expression in corticospinal neurons. Finally, intracortical microstimulation induces activation of only contralateral forelimb muscles in control mice, whereas it induces activation of both contralateral and ipsilateral muscles in mice with cortical inhibition, suggesting that the ipsilaterally projecting CS axons that have been maintained in mice with cortical inhibition form functional connections. Together, these results provide evidence of a potential link between the repellent signaling of Sema6D-PlexA1 and neuronal activity to regulate axon elimination.SIGNIFICANCE STATEMENT Both axon guidance molecules and neuronal activity regulate axon elimination to refine neuronal circuits during development. However, the degree to which these mechanisms operate independently or cooperatively to guide network generation is unclear. Here, we show that neuronal activity-driven Bax/Bak-caspase signaling induces expression of the PlexA1 receptor for the repellent Sema6D molecule in corticospinal neurons (CSNs). This cascade eliminates ipsilateral projections of CSNs in the spinal cord during early postnatal development. The absence of PlexA1, neuronal activity, Bax and Bak, or caspase-9 leads to the maintenance of ipsilateral projections of CSNs, which can form functional connections with spinal neurons. Together, these studies reveal how the Sema6D-PlexA1 signaling and neuronal activity may play a cooperative role in refining CS axonal projections.
Collapse
|
26
|
Jiang T, Zhang G, Liang Y, Cai Z, Liang Z, Lin H, Tan M. PlexinA3 Interacts with CRMP2 to Mediate Sema3A Signalling During Dendritic Growth in Cultured Cerebellar Granule Neurons. Neuroscience 2020; 434:83-92. [DOI: 10.1016/j.neuroscience.2020.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 11/26/2022]
|
27
|
FARP1 boosts CDC42 activity from integrin αvβ5 signaling and correlates with poor prognosis of advanced gastric cancer. Oncogenesis 2020; 9:13. [PMID: 32029704 PMCID: PMC7005035 DOI: 10.1038/s41389-020-0190-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 12/11/2019] [Accepted: 01/10/2020] [Indexed: 02/01/2023] Open
Abstract
Considering the poor prognosis of most advanced cancers, prevention of invasion and metastasis is essential for disease control. Ras homologous (Rho) guanine exchange factors (GEFs) and their signaling cascade could be potential therapeutic targets in advanced cancers. We conducted in silico analyses of The Cancer Genome Atlas expression data to identify candidate Rho-GEF genes showing aberrant expression in advanced gastric cancer and found FERM, Rho/ArhGEF, and pleckstrin domain protein 1 (FARP1) expression is related to poor prognosis. Analyses in 91 clinical advanced gastric cancers of the relationship of prognosis and pathological factors with immunohistochemical expression of FARP1 indicated that high expression of FARP1 is significantly associated with lymphatic invasion, lymph metastasis, and poor prognosis of the patients (P = 0.025). In gastric cancer cells, FARP1 knockdown decreased cell motility, whereas FARP1 overexpression promoted cell motility and filopodium formation via CDC42 activation. FARP1 interacted with integrin β5, and a potent integrin αvβ5 inhibitor (SB273005) prevented cell motility in only high FARP1-expressing gastric cancer cells. These results suggest that the integrin αvβ5-FARP1-CDC42 axis plays a crucial role in gastric cancer cell migration and invasion. Thus, regulatory cascade upstream of Rho can be a specific and promising target of advanced cancer treatment.
Collapse
|
28
|
Zhou X, Bessereau JL. Molecular Architecture of Genetically-Tractable GABA Synapses in C. elegans. Front Mol Neurosci 2019; 12:304. [PMID: 31920535 PMCID: PMC6920096 DOI: 10.3389/fnmol.2019.00304] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Inhibitory synapses represent a minority of the total chemical synapses in the mammalian brain, yet proper tuning of inhibition is fundamental to shape neuronal network properties. The neurotransmitter γ-aminobutyric acid (GABA) mediates rapid synaptic inhibition by the activation of the type A GABA receptor (GABAAR), a pentameric chloride channel that governs major inhibitory neuronal transduction in the nervous system. Impaired GABA transmission leads to a variety of neuropsychiatric diseases, including schizophrenia, autism, epilepsy or anxiety. From an evolutionary perspective, GABAAR shows remarkable conservations, and are found in all eukaryotic clades and even in bacteria and archaea. Specifically, bona fide GABAARs are found in the nematode Caenorhabditis elegans. Because of the anatomical simplicity of the nervous system and its amenability to genetic manipulations, C. elegans provide a powerful system to investigate the molecular and cellular biology of GABA synapses. In this mini review article, we will introduce the structure of the C. elegans GABAergic system and describe recent advances that have identified novel proteins controlling the localization of GABAARs at synapses. In particular, Ce-Punctin/MADD-4 is an evolutionarily-conserved extracellular matrix protein that behaves as an anterograde synaptic organizer to instruct the excitatory or inhibitory identity of postsynaptic domains.
Collapse
Affiliation(s)
- Xin Zhou
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Jean-Louis Bessereau
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| |
Collapse
|
29
|
The Nebulin Family LIM and SH3 Proteins Regulate Postsynaptic Development and Function. J Neurosci 2019; 40:526-541. [PMID: 31754010 PMCID: PMC6961999 DOI: 10.1523/jneurosci.0334-19.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
Neuronal dendrites have specialized actin-rich structures called dendritic spines that receive and integrate most excitatory synaptic inputs. The stabilization of dendrites and spines during neuronal maturation is essential for proper neural circuit formation. Changes in dendritic morphology and stability are largely mediated by regulation of the actin cytoskeleton; however, the underlying mechanisms remain to be fully elucidated. Here, we present evidence that the nebulin family members LASP1 and LASP2 play an important role in the postsynaptic development of rat hippocampal neurons from both sexes. We find that both LASP1 and LASP2 are enriched in dendritic spines, and their knockdown impairs spine development and synapse formation. Furthermore, LASP2 exerts a distinct role in dendritic arbor and dendritic spine stabilization. Importantly, the actin-binding N-terminal LIM domain and nebulin repeats of LASP2 are required for spine stability and dendritic arbor complexity. These findings identify LASP1 and LASP2 as novel regulators of neuronal circuitry.SIGNIFICANCE STATEMENT Proper regulation of the actin cytoskeleton is essential for the structural stability of dendrites and dendritic spines. Consequently, the malformation of dendritic structures accompanies numerous neurologic disorders, such as schizophrenia and autism. Nebulin family members are best known for their role in regulating the stabilization and function of actin thin filaments in muscle. The two smallest family members, LASP1 and LASP2, are more structurally diverse and are expressed in a broader array of tissues. While both LASP1 and LASP2 are highly expressed in the brain, little is currently known about their function in the nervous system. In this study, we demonstrate the first evidence that LASP1 and LASP2 are involved in the formation and long-term maintenance of dendrites and dendritic spines.
Collapse
|
30
|
Libanje F, Raingeaud J, Luan R, Thomas Z, Zajac O, Veiga J, Marisa L, Adam J, Boige V, Malka D, Goéré D, Hall A, Soazec J, Prall F, Gelli M, Dartigues P, Jaulin F. ROCK2 inhibition triggers the collective invasion of colorectal adenocarcinomas. EMBO J 2019; 38:e99299. [PMID: 31304629 PMCID: PMC6627234 DOI: 10.15252/embj.201899299] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/19/2019] [Accepted: 05/10/2019] [Indexed: 12/20/2022] Open
Abstract
The metastatic progression of cancer is a multi-step process initiated by the local invasion of the peritumoral stroma. To identify the mechanisms underlying colorectal carcinoma (CRC) invasion, we collected live human primary cancer specimens at the time of surgery and monitored them ex vivo. This revealed that conventional adenocarcinomas undergo collective invasion while retaining their epithelial glandular architecture with an inward apical pole delineating a luminal cavity. To identify the underlying mechanisms, we used microscopy-based assays on 3D organotypic cultures of Caco-2 cysts as a model system. We performed two siRNA screens targeting Rho-GTPases effectors and guanine nucleotide exchange factors. These screens revealed that ROCK2 inhibition triggers the initial leader/follower polarization of the CRC cell cohorts and induces collective invasion. We further identified FARP2 as the Rac1 GEF necessary for CRC collective invasion. However, FARP2 activation is not sufficient to trigger leader cell formation and the concomitant inhibition of Myosin-II is required to induce invasion downstream of ROCK2 inhibition. Our results contrast with ROCK pro-invasive function in other cancers, stressing that the molecular mechanism of metastatic spread likely depends on tumour types and invasion mode.
Collapse
Affiliation(s)
| | | | - Rui Luan
- INSERM U‐981Gustave RoussyVillejuifFrance
| | | | - Olivier Zajac
- INSERM U‐981Gustave RoussyVillejuifFrance
- Present address:
Department of Translational ResearchCurie InstituteParisFrance
| | - Joel Veiga
- Cell Biology ProgramMemorial Sloan‐Kettering Cancer CenterNew YorkNYUSA
- Present address:
Imagine InstituteParisFrance
| | - Laetitia Marisa
- Programme “Cartes d'Identité des Tumeurs”Ligue Nationale Contre le CancerParisFrance
| | - Julien Adam
- Pathology DepartmentGustave RoussyVillejuifFrance
| | | | - David Malka
- Digestive Cancer UnitGustave RoussyVillejuifFrance
| | - Diane Goéré
- Digestive Cancer UnitGustave RoussyVillejuifFrance
| | - Alan Hall
- Cell Biology ProgramMemorial Sloan‐Kettering Cancer CenterNew YorkNYUSA
| | | | - Friedrich Prall
- Institute of PathologyUniversity Medicine of RostockRostockGermany
| | | | | | | |
Collapse
|
31
|
FARP1 Facilitates Cell Proliferation Through Modulating MAPK Signaling Pathway in Cutaneous Melanoma. Am J Dermatopathol 2019; 41:908-913. [PMID: 31021836 DOI: 10.1097/dad.0000000000001426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose of our study was to investigate the biological functions of FARP1 gene in cutaneous melanoma. METHODS The mRNA expression level of FARP1 in cutaneous melanoma was analyzed based on the data obtained from ONCOMINE and The Cancer Genome Atlas database. Kaplan-Meier analysis was conducted to explore the association between FARP1 expression and the overall survival time of patients with cutaneous melanoma. The mRNA expression of FARP1 in melanoma cells was determined by qRT-PCR. A-375 cell line with silenced FARP1 was constructed to explore its biological functions. Cell proliferation, migration, and invasion abilities were determined by CCK8 assay, wound-healing assay, and transwell assays, respectively. Western blot was performed to explore the protein expression of FARP1, pMEK, MEK, pERK, and ERK. RESULTS Our results showed that the expression level of FARP1 was upregulated in cutaneous melanoma tissues and cells. Kaplan-Meier analysis revealed that high expression of FARP1 is predictive of shorter overall survival time in patients with cutaneous melanoma. Through CCK8 assay, we found that knockdown of FARP1 in A-375 cells exhibited dramatically inhibitory effect on cell proliferation. The results of wound-healing and transwell assays revealed that the motility of A-375 cells was notably suppressed after silencing FARP1. Moreover, the relative expression levels of pMEK/MEK and pERK/ERK decreased remarkably in A-375 cells following being transfected with si-FARP1. CONCLUSIONS Our present results preliminary proofed that FARP1 possibly acts as a promoter in cutaneous melanoma development and possesses the potential to be a therapeutic target in patients with cutaneous melanoma.
Collapse
|
32
|
Xu Z, Fang P, Xu B, Lu Y, Xiong J, Gao F, Wang X, Fan J, Shi P. High-throughput three-dimensional chemotactic assays reveal steepness-dependent complexity in neuronal sensation to molecular gradients. Nat Commun 2018; 9:4745. [PMID: 30420609 PMCID: PMC6232128 DOI: 10.1038/s41467-018-07186-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/19/2018] [Indexed: 01/28/2023] Open
Abstract
Many cellular programs of neural development are under combinatorial regulation by different chemoattractive or chemorepulsive factors. Here, we describe a microfluidic platform that utilizes well-controlled three-dimensional (3D) diffusion to generate molecular gradients of varied steepness in a large array of hydrogel cylinders, allowing high-throughput 3D chemotactic assays for mechanistic dissection of steepness-dependent neuronal chemotaxis. Using this platform, we examine neuronal sensitivity to the steepness of gradient composed of netrin-1, nerve growth factor, or semaphorin3A (Sema3A) proteins, and reveal dramatic diversity and complexity in the associated chemotactic regulation of neuronal development. Particularly for Sema3A, we find that serine/threonine kinase-11 and glycogen synthase kinase-3 signaling pathways are differentially involved in steepness-dependent chemotactic regulation of coordinated neurite repellence and neuronal migration. These results provide insights to the critical role of gradient steepness in neuronal chemotaxis, and also prove the technique as an expandable platform for studying other chemoresponsive cellular systems.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China
| | - Peilin Fang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China
| | - Bingzhe Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China
| | - Yufeng Lu
- Department of Material Science and Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China
| | - Jinghui Xiong
- Department of Material Science and Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China
| | - Feng Gao
- Department of Biomedical Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
| | - Xin Wang
- Department of Biomedical Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, 518000, Shenzhen, China
| | - Jun Fan
- Department of Material Science and Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Ave, Kowloon, Hong Kong SAR, 999077, China.
- Shenzhen Research Institute, City University of Hong Kong, 518000, Shenzhen, China.
| |
Collapse
|
33
|
Kuo YC, He X, Coleman AJ, Chen YJ, Dasari P, Liou J, Biederer T, Zhang X. Structural analyses of FERM domain-mediated membrane localization of FARP1. Sci Rep 2018; 8:10477. [PMID: 29992992 PMCID: PMC6041286 DOI: 10.1038/s41598-018-28692-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
FARP1 is a multi-domain protein that is involved in regulating neuronal development through interacting with cell surface proteins such as class A Plexins and SynCAM 1. The N-terminal FERM domain in FARP1 is known to both promote membrane localization and mediate these protein interactions, for which the underlying molecular mechanisms remain unclear. Here we determined the crystal structures of the FERM domain of FARP1 from zebrafish, and those of FARP2 (a close homolog of FARP1) from mouse and zebrafish. These FERM domains adopt the three-leaved clover fold that is typical of all FERM domains. Our structures reveal a positively charged surface patch that is highly conserved in the FERM domain of FARP1 and FARP2. In vitro lipid-binding experiments showed that the FARP1 FERM domain binds specifically to several types of phospholipid, which is dependent on the positively charged surface patch. We further determined through cell-based analyses that this surface patch on the FERM domain underlies the localization of FARP1 to the plasma membrane, and that FERM domain interactions recruit it to postsynaptic sites in neurons.
Collapse
Affiliation(s)
- Yi-Chun Kuo
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xiaojing He
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Andrew J Coleman
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Yu-Ju Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pranathi Dasari
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Xuewu Zhang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
34
|
Shadrach JL, Pierchala BA. Semaphorin3A Signaling Is Dispensable for Motor Axon Reinnervation of the Adult Neuromuscular Junction. eNeuro 2018; 5:ENEURO.0155-17.2018. [PMID: 29774231 PMCID: PMC5955010 DOI: 10.1523/eneuro.0155-17.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 04/23/2018] [Accepted: 04/27/2018] [Indexed: 01/13/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized synapse that is formed by motor axon innervation of skeletal muscle fibers. The maintenance of motor-muscle connectivity is critical for the preservation of muscle tone and generation of movement. Injury can induce a robust regenerative response in motor axons, but severe trauma or chronic denervation resulting from neurodegenerative disease typically leads to inefficient repair and poor functional recovery. The axon guidance molecule Semaphorin3A (Sema3A) has been implicated as a negative regulator of motor innervation. Upon binding to a plexinA-neuropilin1 (Npn1) receptor complex, Sema3A initiates a downstream signaling cascade that results in axonal repulsion. Here, we established a reproducible nerve crush model to quantify motor nerve regeneration. We then used that model to investigate the role of Sema3A signaling at the adult NMJ. In contrast to previous findings, we found that Sema3A and Npn1 mRNA decrease in response to denervation, suggesting that Sema3A-Npn1 signaling may regulate NMJ reinnervation. To directly test that hypothesis, we used inducible knockout models to ubiquitously delete Sema3A or Npn1 from adult mice. Despite demonstrating that we could achieve highly efficient gene deletion, disruption of Sema3A-Npn1 signaling did not affect the normal maintenance of the NMJ or disrupt motor axon reinnervation after a denervating injury.
Collapse
Affiliation(s)
- Jennifer L. Shadrach
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109
- Program in Cellular and Molecular Biology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109
| | - Brian A. Pierchala
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan 48109
- Program in Cellular and Molecular Biology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109
| |
Collapse
|
35
|
Spurlin JW, Nelson CM. Building branched tissue structures: from single cell guidance to coordinated construction. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2015.0527. [PMID: 28348257 DOI: 10.1098/rstb.2015.0527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
Branched networks are ubiquitous throughout nature, particularly found in tissues that require large surface area within a restricted volume. Many tissues with a branched architecture, such as the vasculature, kidney, mammary gland, lung and nervous system, function to exchange fluids, gases and information throughout the body of an organism. The generation of branched tissues requires regulation of branch site specification, initiation and elongation. Branching events often require the coordination of many cells to build a tissue network for material exchange. Recent evidence has emerged suggesting that cell cooperativity scales with the number of cells actively contributing to branching events. Here, we compare mechanisms that regulate branching, focusing on how cell cohorts behave in a coordinated manner to build branched tissues.This article is part of the themed issue 'Systems morphodynamics: understanding the development of tissue hardware'.
Collapse
Affiliation(s)
- James W Spurlin
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA .,Molecular Biology, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| |
Collapse
|
36
|
Salzberg Y, Coleman AJ, Celestrin K, Cohen-Berkman M, Biederer T, Henis-Korenblit S, Bülow HE. Reduced Insulin/Insulin-Like Growth Factor Receptor Signaling Mitigates Defective Dendrite Morphogenesis in Mutants of the ER Stress Sensor IRE-1. PLoS Genet 2017; 13:e1006579. [PMID: 28114319 PMCID: PMC5293268 DOI: 10.1371/journal.pgen.1006579] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/06/2017] [Accepted: 01/11/2017] [Indexed: 11/18/2022] Open
Abstract
Neurons receive excitatory or sensory inputs through their dendrites, which often branch extensively to form unique neuron-specific structures. How neurons regulate the formation of their particular arbor is only partially understood. In genetic screens using the multidendritic arbor of PVD somatosensory neurons in the nematode Caenorhabditis elegans, we identified a mutation in the ER stress sensor IRE-1/Ire1 (inositol requiring enzyme 1) as crucial for proper PVD dendrite arborization in vivo. We further found that regulation of dendrite growth in cultured rat hippocampal neurons depends on Ire1 function, showing an evolutionarily conserved role for IRE-1/Ire1 in dendrite patterning. PVD neurons of nematodes lacking ire-1 display reduced arbor complexity, whereas mutations in genes encoding other ER stress sensors displayed normal PVD dendrites, specifying IRE-1 as a selective ER stress sensor that is essential for PVD dendrite morphogenesis. Although structure function analyses indicated that IRE-1's nuclease activity is necessary for its role in dendrite morphogenesis, mutations in xbp-1, the best-known target of non-canonical splicing by IRE-1/Ire1, do not exhibit PVD phenotypes. We further determined that secretion and distal localization to dendrites of the DMA-1/leucine rich transmembrane receptor (DMA-1/LRR-TM) is defective in ire-1 but not xbp-1 mutants, suggesting a block in the secretory pathway. Interestingly, reducing Insulin/IGF1 signaling can bypass the secretory block and restore normal targeting of DMA-1, and consequently normal PVD arborization even in the complete absence of functional IRE-1. This bypass of ire-1 requires the DAF-16/FOXO transcription factor. In sum, our work identifies a conserved role for ire-1 in neuronal branching, which is independent of xbp-1, and suggests that arborization defects associated with neuronal pathologies may be overcome by reducing Insulin/IGF signaling and improving ER homeostasis and function.
Collapse
Affiliation(s)
- Yehuda Salzberg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Andrew J. Coleman
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States of America
| | - Kevin Celestrin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Moran Cohen-Berkman
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States of America
| | - Sivan Henis-Korenblit
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Hannes E. Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
37
|
Xu C, Fu X, Zhu S, Liu JJ. Retrolinkin recruits the WAVE1 protein complex to facilitate BDNF-induced TrkB endocytosis and dendrite outgrowth. Mol Biol Cell 2016; 27:3342-3356. [PMID: 27605705 PMCID: PMC5170866 DOI: 10.1091/mbc.e16-05-0326] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022] Open
Abstract
Retrolinkin, a neuronal membrane protein, coordinates with endophilin A1 and mediates early endocytic trafficking and signal transduction of the ligand-receptor complex formed between brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), in dendrites of CNS neurons. Here we report that retrolinkin interacts with the CYFIP1/2 subunit of the WAVE1 complex, a member of the WASP/WAVE family of nucleation-promoting factors that binds and activates the Arp2/3 complex to promote branched actin polymerization. WAVE1, not N-WASP, is required for BDNF-induced TrkB endocytosis and dendrite outgrowth. Disruption of the interaction between retrolinkin and CYFIP1/2 impairs recruitment of WAVE1 to neuronal plasma membrane upon BDNF addition and blocks internalization of activated TrkB. We also show that WAVE1-mediated endocytosis of BDNF-activated TrkB is actin dependent and clathrin independent. These results not only reveal the mechanistic role of retrolinkin in BDNF-TrkB endocytosis, but also indicate that WASP/WAVE-dependent actin polymerization during endocytosis is regulated by cell type-specific and cargo-specific modulators.
Collapse
Affiliation(s)
- Chenchang Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China.,Graduate School, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Xiuping Fu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China.,Graduate School, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Shaoxia Zhu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jia-Jia Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
38
|
Schwaid AG, Su C, Loos P, Wu J, Nguyen C, Stone KL, Kanyo J, Geoghegan KF, Bhattacharya SK, Dow RL, Buckbinder L, Carpino PA. MAP4K4 Is a Threonine Kinase That Phosphorylates FARP1. ACS Chem Biol 2015; 10:2667-71. [PMID: 26422651 DOI: 10.1021/acschembio.5b00679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mitogen-activated protein kinase 4 (MAP4K4) regulates the MEK kinase cascade and is implicated in cytoskeletal rearrangement and migration; however, identifying MAP4K4 substrates has remained a challenge. To ascertain MAP4K4-dependent phosphorylation events, we combined phosphoproteomic studies of MAP4K4 inhibition with in vitro assessment of its kinase specificity. We identified 235 phosphosites affected by MAP4K4 inhibition in cells and found that pTP and pSP motifs were predominant among them. In contrast, in vitro assessment of kinase specificity showed that MAP4K4 favors a pTL motif. We showed that MAP4K4 directly phosphorylates and coimmunoprecipitates with FERM, RhoGEF, and pleckstrin domain-containing protein 1 (FARP1). MAP4K4 inhibition in SH-SY5Y cells increases neurite outgrowth, a process known to involve FARP1. As FARP1 and MAP4K4 both contribute to cytoskeletal rearrangement, the results suggest that MAP4K4 exerts some of its effects on the cytoskeleton via phosphorylation of FARP1.
Collapse
Affiliation(s)
| | | | | | - Jiang Wu
- Structural
Biology and Biophysics, Center for Chemistry Innovation and Excellence, Pfizer Pharmatherapeutics Research and Development, Groton, Connecticut 06340, United States
| | - Chuong Nguyen
- Structural
Biology and Biophysics, Center for Chemistry Innovation and Excellence, Pfizer Pharmatherapeutics Research and Development, Groton, Connecticut 06340, United States
| | - Kathryn L. Stone
- W.M.
Keck Foundation Biotechnology Resource Laboratory, Yale University, New Haven, Connecticut 06520, United States
| | - Jean Kanyo
- W.M.
Keck Foundation Biotechnology Resource Laboratory, Yale University, New Haven, Connecticut 06520, United States
| | - Kieran F. Geoghegan
- Structural
Biology and Biophysics, Center for Chemistry Innovation and Excellence, Pfizer Pharmatherapeutics Research and Development, Groton, Connecticut 06340, United States
| | | | | | | | | |
Collapse
|
39
|
Yamashita N, Aoki R, Chen S, Jitsuki-Takahashi A, Ohura S, Kamiya H, Goshima Y. Voltage-gated calcium and sodium channels mediate Sema3A retrograde signaling that regulates dendritic development. Brain Res 2015; 1631:127-36. [PMID: 26638837 DOI: 10.1016/j.brainres.2015.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 10/22/2022]
Abstract
Growing axons rely on local signaling at the growth cone for guidance cues. Semaphorin3A (Sema3A), a secreted repulsive axon guidance molecule, regulates synapse maturation and dendritic branching. We previously showed that local Sema3A signaling in the growth cones elicits retrograde retrograde signaling via PlexinA4 (PlexA4), one component of the Sema3A receptor, thereby regulating dendritic localization of AMPA receptor GluA2 and proper dendritic development. In present study, we found that nimodipine (voltage-gated L-type Ca(2+) channel blocker) and tetrodotoxin (TTX; voltage-gated Na(+) channel blocker) suppress Sema3A-induced dendritic localization of GluA2 and dendritic branch formation in cultured hippocampal neurons. The local application of nimodipine or TTX to distal axons suppresses retrograde transport of Venus-Sema3A that has been exogenously applied to the distal axons. Sema3A facilitates axonal transport of PlexA4, which is also suppressed in neurons treated with either TTX or nimodipine. These data suggest that voltage-gated calcium and sodium channels mediate Sema3A retrograde signaling that regulates dendritic GluA2 localization and branch formation.
Collapse
Affiliation(s)
- Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA; JSPS Postdoctoral Fellowship for Research Abroad, Chiyoda-ku 102-0083, Japan
| | - Reina Aoki
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Sandy Chen
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Aoi Jitsuki-Takahashi
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Shunsuke Ohura
- Department of Neurobiology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Haruyuki Kamiya
- Department of Neurobiology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.
| |
Collapse
|
40
|
Membrane and Protein Interactions of the Pleckstrin Homology Domain Superfamily. MEMBRANES 2015; 5:646-63. [PMID: 26512702 PMCID: PMC4704004 DOI: 10.3390/membranes5040646] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 12/23/2022]
Abstract
The human genome encodes about 285 proteins that contain at least one annotated pleckstrin homology (PH) domain. As the first phosphoinositide binding module domain to be discovered, the PH domain recruits diverse protein architectures to cellular membranes. PH domains constitute one of the largest protein superfamilies, and have diverged to regulate many different signaling proteins and modules such as Dbl homology (DH) and Tec homology (TH) domains. The ligands of approximately 70 PH domains have been validated by binding assays and complexed structures, allowing meaningful extrapolation across the entire superfamily. Here the Membrane Optimal Docking Area (MODA) program is used at a genome-wide level to identify all membrane docking PH structures and map their lipid-binding determinants. In addition to the linear sequence motifs which are employed for phosphoinositide recognition, the three dimensional structural features that allow peripheral membrane domains to approach and insert into the bilayer are pinpointed and can be predicted ab initio. The analysis shows that conserved structural surfaces distinguish which PH domains associate with membrane from those that do not. Moreover, the results indicate that lipid-binding PH domains can be classified into different functional subgroups based on the type of membrane insertion elements they project towards the bilayer.
Collapse
|
41
|
Li Q, Zhang Y, Zou J, Qi F, Yang J, Yuan Q, Yao Z. Neonatal vaccination with bacille Calmette-Guérin promotes the dendritic development of hippocampal neurons. Hum Vaccin Immunother 2015; 12:140-9. [PMID: 26375414 DOI: 10.1080/21645515.2015.1056954] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Dendritic structure is sensitive to changes in the environment during brain development. Accumulating evidence has demonstrated that early immune activation can significantly affect neuronal development. Our study concentrated on the morphological study of neural dendrites and spines in the hippocampal CA1 area using Diolistic labeling with Sholl analysis and fractal analysis. The results revealed that Bacille Calmette-Guérin (BCG) vaccination enhanced dendritic complexity, as reflected by the increased number of intersections, number of branch points and fractal dimension, and promoted neurite outgrowth. In addition, BCG increased the density and promoted the maturation of dendritic spines. The alterations in dendritic structure and spine morphology were observed at 2 and 4 w, but the differences were more apparent at 4 w than at 2 w. However, no significant difference was observed at 8 w. Furthermore, we observed that BCG increased the expression of hippocampal brain derived neurotrophic factor (BDNF) and insulin-like growth factor 1 (IGF-1). Hippocampal BDNF/IGF-1 was positively correlated with apical dendritic length, fractal dimension, and spine density. Taken together, we show in this study that neonatal BCG vaccination promotes dendritic development in developing hippocampal CA1 neurons, most likely by increasing the expression of BDNF and IGF-1 in the hippocampus.
Collapse
Affiliation(s)
- Qingqing Li
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Yuwei Zhang
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Juntao Zou
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Fangfang Qi
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Junhua Yang
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Qunfang Yuan
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Zhibin Yao
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| |
Collapse
|
42
|
Regulating Rac in the nervous system: molecular function and disease implication of Rac GEFs and GAPs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:632450. [PMID: 25879033 PMCID: PMC4388020 DOI: 10.1155/2015/632450] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/06/2015] [Indexed: 12/11/2022]
Abstract
Rho family GTPases, including RhoA, Rac1, and Cdc42 as the most studied members, are master regulators of actin cytoskeletal organization. Rho GTPases control various aspects of the nervous system and are associated with a number of neuropsychiatric and neurodegenerative diseases. The activity of Rho GTPases is controlled by two families of regulators, guanine nucleotide exchange factors (GEFs) as the activators and GTPase-activating proteins (GAPs) as the inhibitors. Through coordinated regulation by GEFs and GAPs, Rho GTPases act as converging signaling molecules that convey different upstream signals in the nervous system. So far, more than 70 members of either GEFs or GAPs of Rho GTPases have been identified in mammals, but only a small subset of them have well-known functions. Thus, characterization of important GEFs and GAPs in the nervous system is crucial for the understanding of spatiotemporal dynamics of Rho GTPase activity in different neuronal functions. In this review, we summarize the current understanding of GEFs and GAPs for Rac1, with emphasis on the molecular function and disease implication of these regulators in the nervous system.
Collapse
|