1
|
Nicolas-Martinez EC, Robinson O, Pflueger C, Gardner A, Corbett MA, Ritchie T, Kroes T, van Eyk CL, Scheffer IE, Hildebrand MS, Barnier JV, Rousseau V, Genevieve D, Haushalter V, Piton A, Denommé-Pichon AS, Bruel AL, Nambot S, Isidor B, Grigg J, Gonzalez T, Ghedia S, Marchant RG, Bournazos A, Wong WK, Webster RI, Evesson FJ, Jones KJ, Cooper ST, Lister R, Gecz J, Jolly LA. RNA variant assessment using transactivation and transdifferentiation. Am J Hum Genet 2024; 111:1673-1699. [PMID: 39084224 PMCID: PMC11339655 DOI: 10.1016/j.ajhg.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Understanding the impact of splicing and nonsense variants on RNA is crucial for the resolution of variant classification as well as their suitability for precision medicine interventions. This is primarily enabled through RNA studies involving transcriptomics followed by targeted assays using RNA isolated from clinically accessible tissues (CATs) such as blood or skin of affected individuals. Insufficient disease gene expression in CATs does however pose a major barrier to RNA based investigations, which we show is relevant to 1,436 Mendelian disease genes. We term these "silent" Mendelian genes (SMGs), the largest portion (36%) of which are associated with neurological disorders. We developed two approaches to induce SMG expression in human dermal fibroblasts (HDFs) to overcome this limitation, including CRISPR-activation-based gene transactivation and fibroblast-to-neuron transdifferentiation. Initial transactivation screens involving 40 SMGs stimulated our development of a highly multiplexed transactivation system culminating in the 6- to 90,000-fold induction of expression of 20/20 (100%) SMGs tested in HDFs. Transdifferentiation of HDFs directly to neurons led to expression of 193/516 (37.4%) of SMGs implicated in neurological disease. The magnitude and isoform diversity of SMG expression following either transactivation or transdifferentiation was comparable to clinically relevant tissues. We apply transdifferentiation and/or gene transactivation combined with short- and long-read RNA sequencing to investigate the impact that variants in USH2A, SCN1A, DMD, and PAK3 have on RNA using HDFs derived from affected individuals. Transactivation and transdifferentiation represent rapid, scalable functional genomic solutions to investigate variants impacting SMGs in the patient cell and genomic context.
Collapse
Affiliation(s)
- Emmylou C Nicolas-Martinez
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Olivia Robinson
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Christian Pflueger
- Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; Australian Research Council Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Alison Gardner
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Mark A Corbett
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Tarin Ritchie
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Thessa Kroes
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Clare L van Eyk
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia; Murdoch Children's Research Institute, Parkville, VIC 3052, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, VIC 3052, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jean-Vianney Barnier
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Véronique Rousseau
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - David Genevieve
- Montpellier University, Inserm U1183, Reference Center for Rare Diseases Developmental Anomaly and Malformative Syndromes, Genetics Department, Montpellier Hospital, Montpellier, France
| | - Virginie Haushalter
- Genetic Diagnosis Laboratory, Strasbourg University Hospital, Strasbourg, France
| | - Amélie Piton
- Genetic Diagnosis Laboratory, Strasbourg University Hospital, Strasbourg, France
| | - Anne-Sophie Denommé-Pichon
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France; INSERM UMR1231, GAD "Génétique des Anomalies du Développement," FHU-TRANSLAD, University of Burgundy, Dijon, France
| | - Ange-Line Bruel
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France; INSERM UMR1231, GAD "Génétique des Anomalies du Développement," FHU-TRANSLAD, University of Burgundy, Dijon, France
| | - Sophie Nambot
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France; INSERM UMR1231, GAD "Génétique des Anomalies du Développement," FHU-TRANSLAD, University of Burgundy, Dijon, France
| | - Bertrand Isidor
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France; INSERM UMR1231, GAD "Génétique des Anomalies du Développement," FHU-TRANSLAD, University of Burgundy, Dijon, France
| | - John Grigg
- Speciality of Ophthalmology, Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia
| | - Tina Gonzalez
- Department of Clinical Genetics, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Sondhya Ghedia
- Department of Clinical Genetics, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Rhett G Marchant
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia
| | - Adam Bournazos
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Wui-Kwan Wong
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Children's Medical Research Institute, Westmead, NSW 2145, Australia; Department of Paediatric Neurology, Children's Hospital at Westmead, Sydney, NSW 2000, Australia
| | - Richard I Webster
- Department of Paediatric Neurology, Children's Hospital at Westmead, Sydney, NSW 2000, Australia
| | - Frances J Evesson
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Kristi J Jones
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Children's Medical Research Institute, Westmead, NSW 2145, Australia; Department of Clinical Genetics, Children's Hospital at Westmead, Sydney, NSW 2000, Australia
| | - Sandra T Cooper
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; Children's Medical Research Institute, Westmead, NSW 2145, Australia
| | - Ryan Lister
- Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; Australian Research Council Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Jozef Gecz
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia.
| | - Lachlan A Jolly
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia; School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
2
|
Dobrigna M, Poëa-Guyon S, Rousseau V, Vincent A, Toutain A, Barnier JV. The molecular basis of p21-activated kinase-associated neurodevelopmental disorders: From genotype to phenotype. Front Neurosci 2023; 17:1123784. [PMID: 36937657 PMCID: PMC10017488 DOI: 10.3389/fnins.2023.1123784] [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: 12/14/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Although the identification of numerous genes involved in neurodevelopmental disorders (NDDs) has reshaped our understanding of their etiology, there are still major obstacles in the way of developing therapeutic solutions for intellectual disability (ID) and other NDDs. These include extensive clinical and genetic heterogeneity, rarity of recurrent pathogenic variants, and comorbidity with other psychiatric traits. Moreover, a large intragenic mutational landscape is at play in some NDDs, leading to a broad range of clinical symptoms. Such diversity of symptoms is due to the different effects DNA variations have on protein functions and their impacts on downstream biological processes. The type of functional alterations, such as loss or gain of function, and interference with signaling pathways, has yet to be correlated with clinical symptoms for most genes. This review aims at discussing our current understanding of how the molecular changes of group I p21-activated kinases (PAK1, 2 and 3), which are essential actors of brain development and function; contribute to a broad clinical spectrum of NDDs. Identifying differences in PAK structure, regulation and spatio-temporal expression may help understanding the specific functions of each group I PAK. Deciphering how each variation type affects these parameters will help uncover the mechanisms underlying mutation pathogenicity. This is a prerequisite for the development of personalized therapeutic approaches.
Collapse
Affiliation(s)
- Manon Dobrigna
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Sandrine Poëa-Guyon
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Véronique Rousseau
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Aline Vincent
- Department of Genetics, EA7450 BioTARGen, University Hospital of Caen, Caen, France
| | - Annick Toutain
- Department of Genetics, University Hospital of Tours, UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Jean-Vianney Barnier
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
- *Correspondence: Jean-Vianney Barnier,
| |
Collapse
|
3
|
Liu K, Su Q, Kang K, Chen M, Wang WX, Zhang WQ, Pang R. Genome-wide Analysis of Alternative Gene Splicing Associated with Virulence in the Brown Planthopper Nilaparvata lugens (Hemiptera: Delphacidae). JOURNAL OF ECONOMIC ENTOMOLOGY 2021; 114:2512-2523. [PMID: 34568947 DOI: 10.1093/jee/toab186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Indexed: 06/13/2023]
Abstract
Alternative splicing of protein coding genes plays a profound role in phenotypic variation for many eukaryotic organisms. The development of high-throughput sequencing and bioinformatics algorithms provides the possibility of genome-wide identification of alternative splicing events in eukaryotes. However, for the brown planthopper Nilaparvata lugens, a destructive pest of rice crops, whole-genome distribution of alternative splicing events and the role of alternative splicing in the phenotypic plasticity of virulence have not previously been estimated. Here, we developed an analysis pipeline to identify alternative splicing events in the genome of N. lugens. Differential expression analysis and functional annotation were performed on datasets related to different virulence phenotypes. In total, 27,880 alternative splicing events corresponding to 9,787 multi-exon genes were detected in N. lugens. Among them, specifically expressed alternative splicing transcripts in the virulent Mudgo population were enriched in metabolic process categories, while transcripts in the avirulent TN1 population were enriched in regulator activity categories. In addition, genes encoding odorant receptor, secreted saliva protein and xenobiotic metabolic P450 monooxygenase showed different splicing patterns between Mudgo population and TN1 population. Host change experiment also revealed that an isoform of a P450 gene could be specially induced by the stimulation of resistant rice variety Mudgo. This research pioneered a genome-wide study of alternative gene splicing in the rice brown planthopper. Differences in alternative splicing between virulent and avirulent populations indicated that alternative splicing might play an important role in the formation of virulence phenotypes in N. lugens.
Collapse
Affiliation(s)
- Kai Liu
- Innovative Institute for Plant Health, College of Agriculture and Biology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qin Su
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kui Kang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wei-Xia Wang
- State Key Laboratory of Rice Biology, China National Rice Research Institute, Hangzhou, Zhejiang, China
| | - Wen-Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rui Pang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
4
|
Wang Y, Guo F. Group I PAKs in myelin formation and repair of the central nervous system: what, when, and how. Biol Rev Camb Philos Soc 2021; 97:615-639. [PMID: 34811887 DOI: 10.1111/brv.12815] [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: 06/01/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022]
Abstract
p21-activated kinases (PAKs) are a family of cell division control protein 42/ras-related C3 botulinum toxin substrate 1 (Cdc42/Rac1)-activated serine/threonine kinases. Group I PAKs (PAK1-3) have distinct activation mechanisms from group II PAKs (PAK4-6) and are the focus of this review. In transformed cancer cells, PAKs regulate a variety of cellular processes and molecular pathways which are also important for myelin formation and repair in the central nervous system (CNS). De novo mutations in group I PAKs are frequently seen in children with neurodevelopmental defects and white matter anomalies. Group I PAKs regulate virtually every aspect of neuronal development and function. Yet their functions in CNS myelination and remyelination remain incompletely defined. Herein, we highlight the current understanding of PAKs in regulating cellular and molecular pathways and discuss the status of PAK-regulated pathways in oligodendrocyte development. We point out outstanding questions and future directions in the research field of group I PAKs and oligodendrocyte development.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurology, Shriners Hospitals for Children/School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), University of California, Davis, 2425 Stockton Blvd, Sacramento, CA, 95817, U.S.A
| | - Fuzheng Guo
- Department of Neurology, Shriners Hospitals for Children/School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), University of California, Davis, 2425 Stockton Blvd, Sacramento, CA, 95817, U.S.A
| |
Collapse
|
5
|
Disruption of PAK3 Signaling in Social Interaction Induced cFos Positive Cells Impairs Social Recognition Memory. Cells 2021; 10:cells10113010. [PMID: 34831234 PMCID: PMC8616103 DOI: 10.3390/cells10113010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
P21-activated kinase 3 (PAK3) gene mutations are linked to several neurodevelopmental disorders, but the underlying mechanisms remain unclear. In this study, we used a tetracycline-inducible system to control the expression of a mutant PAK3 (mPAK3) protein in immediate early gene, namely cFos, positive cells to disrupt PAK signaling, specifically in cells activated by social interaction in transgenic mice. We show that the expression of mPAK3-GFP proteins was in cFos-expressing excitatory and inhibitory neurons in various brain regions, such as the cortex and hippocampus, commonly activated during learning and memory. Basal expression of mPAK3-GFP proteins in cFos-positive cells resulted in social recognition memory deficits in the three-chamber social interaction test, without affecting locomotor activity or other forms of memory. The social memory deficit was rescued by doxycycline to halt the mPAK3-GFP transgene expression. In addition, we show that the expression of mPAK3-GFP proteins in a subset of cFos-positive cells, induced by an antecedent short social interaction, termed social pairing, was sufficient to impair social recognition memory. These results indicate that normal PAK signaling in cFos-positive cells activated during social interaction is critical for social memory.
Collapse
|
6
|
Li N, Yue L, Wang J, Wan Z, Bu W. MicroRNA-24 alleviates isoflurane-induced neurotoxicity in rat hippocampus via attenuation of oxidative stress. Biochem Cell Biol 2019; 98:208-218. [PMID: 31533001 DOI: 10.1139/bcb-2019-0188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Several miRNAs have been recently suggested as potential therapeutic targets for anesthesia-related diseases. This study was carried out to explore the biological roles of miR-24 in isoflurane-treated rat hippocampal neurons. Isoflurane was used to induce neurotoxicity in a rat model. Gain- and loss-of-function of miR-24 was performed, and the size and Ca2+ permeability of mitochondria, as well as cell proliferation and apoptosis, and levels of oxidative-stress-related factors were measured both in vivo and in vitro. Dual luciferase reporter gene assays were used to identify the target relationship between miR-24 and p27kip1. In this study, isoflurane treatment decreased miR-24 expression, after which, levels of neuron apoptosis and oxidative-stress-related factors were elevated and neuron viability was reduced. Over-expression of miR-24 inhibited oxidative damage and neuronal apoptosis in hippocampal tissues, and suppressed the size and Ca2+ permeability of mitochondria of hippocampal neurons. miR-24 enhanced the viability of rat hippocampal neurons by targeting p27kip1. To conclude, this study demonstrated that miR-24 attenuates isoflurane-induced neurotoxicity in rat hippocampus via its antioxidative properties and inhibiting p27kip1 expression.
Collapse
Affiliation(s)
- Na Li
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China
| | - Linli Yue
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China
| | - Jun Wang
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China.,Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China
| | - Zhenzhen Wan
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China.,Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China
| | - Wenhao Bu
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China.,Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, 430070, P.R. China
| |
Collapse
|
7
|
Lithium Sensitive ORAI1 Expression, Store Operated Ca 2+ Entry and Suicidal Death of Neurons in Chorea-Acanthocytosis. Sci Rep 2017; 7:6457. [PMID: 28743945 PMCID: PMC5526875 DOI: 10.1038/s41598-017-06451-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Chorea-Acanthocytosis (ChAc), a neurodegenerative disorder, results from loss-of-function-mutations of chorein-encoding gene VPS13A. In tumour cells chorein up-regulates ORAI1, a Ca2+-channel accomplishing store operated Ca2+-entry (SOCE) upon stimulation by STIM1. Furthermore SOCE could be up-regulated by lithium. The present study explored whether SOCE impacts on neuron apoptosis. Cortical neurons were differentiated from induced pluripotent stem cells generated from fibroblasts of ChAc patients and healthy volunteers. ORAI1 and STIM1 transcript levels and protein abundance were estimated from qRT-PCR and Western blotting, respectively, cytosolic Ca2+-activity ([Ca2+]i) from Fura-2-fluorescence, as well as apoptosis from annexin-V-binding and propidium-iodide uptake determined by flow cytometry. As a result, ORAI1 and STIM1 transcript levels and protein abundance and SOCE were significantly smaller and the percentage apoptotic cells significantly higher in ChAc neurons than in control neurons. Lithium treatment (2 mM, 24 hours) increased significantly ORAI1 and STIM1 transcript levels and protein abundance, an effect reversed by inhibition of Serum & Glucocorticoid inducible Kinase 1. ORAI1 blocker 2-APB (50 µM, 24 hours) significantly decreased SOCE, markedly increased apoptosis and abrogated the anti-apoptotic effect of lithium. In conclusion, enhanced neuronal apoptosis in ChAc at least partially results from decreased ORAI1 expression and SOCE, which could be reversed by lithium treatment.
Collapse
|
8
|
Franchi SA, Astro V, Macco R, Tonoli D, Barnier JV, Botta M, de Curtis I. Identification of a Protein Network Driving Neuritogenesis of MGE-Derived GABAergic Interneurons. Front Cell Neurosci 2016; 10:289. [PMID: 28066185 PMCID: PMC5174131 DOI: 10.3389/fncel.2016.00289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/02/2016] [Indexed: 12/27/2022] Open
Abstract
Interneurons are essential modulators of brain activity and their abnormal maturation may lead to neural and intellectual disabilities. Here we show that cultures derived from murine medial ganglionic eminences (MGEs) produce virtually pure, polarized γ-aminobutyric acid (GABA)-ergic interneurons that can form morphologically identifiable inhibitory synapses. We show that Rac GTPases and a protein complex including the GIT family scaffold proteins are expressed during maturation in vitro, and are required for the normal development of neurites. GIT1 promotes neurite extension in a conformation-dependent manner, while affecting its interaction with specific partners reduces neurite branching. Proteins of the GIT network are concentrated at growth cones, and interaction mutants may affect growth cone behavior. Our findings identify the PIX/GIT1/liprin-α1/ERC1 network as critical for the regulation of interneuron neurite differentiation in vitro, and show that these cultures represent a valuable system to identify the molecular mechanisms driving the maturation of cortical/hippocampal interneurons.
Collapse
Affiliation(s)
- Sira A Franchi
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute and San Raffaele University Milano, Italy
| | - Veronica Astro
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute and San Raffaele University Milano, Italy
| | - Romina Macco
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute and San Raffaele University Milano, Italy
| | - Diletta Tonoli
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute and San Raffaele University Milano, Italy
| | - Jean-Vianney Barnier
- Neuroscience Paris-Saclay Institute, UMR 9197, Centre National de la Recherche Scientifique-Université Paris-Sud Orsay, France
| | - Martina Botta
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute and San Raffaele University Milano, Italy
| | - Ivan de Curtis
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute and San Raffaele University Milano, Italy
| |
Collapse
|
9
|
Maglorius Renkilaraj MRL, Baudouin L, Wells CM, Doulazmi M, Wehrlé R, Cannaya V, Bachelin C, Barnier JV, Jia Z, Nait Oumesmar B, Dusart I, Bouslama-Oueghlani L. The intellectual disability protein PAK3 regulates oligodendrocyte precursor cell differentiation. Neurobiol Dis 2016; 98:137-148. [PMID: 27940202 DOI: 10.1016/j.nbd.2016.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 11/04/2016] [Accepted: 12/02/2016] [Indexed: 10/20/2022] Open
Abstract
Oligodendrocyte and myelin deficits have been reported in mental/psychiatric diseases. The p21-activated kinase 3 (PAK3), a serine/threonine kinase, whose activity is stimulated by the binding of active Rac and Cdc42 GTPases is affected in these pathologies. Indeed, many mutations of Pak3 gene have been described in non-syndromic intellectual disability diseases. Pak3 is expressed mainly in the brain where its role has been investigated in neurons but not in glial cells. Here, we showed that PAK3 is highly expressed in oligodendrocyte precursors (OPCs) and its expression decreases in mature oligodendrocytes. In the developing white matter of the Pak3 knockout mice, we found defects of oligodendrocyte differentiation in the corpus callosum and to a lesser extent in the anterior commissure, which were compensated at the adult stage. In vitro experiments in OPC cultures, derived from Pak3 knockout and wild type brains, support a developmental and cell-autonomous role for PAK3 in regulating OPC differentiation into mature oligodendrocytes. Moreover, we did not detect any obvious alterations of the proliferation or migration of Pak3 null OPCs compared to wild type. Overall, our data highlight PAK3 as a new regulator of OPC differentiation.
Collapse
Affiliation(s)
| | - Lucas Baudouin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, F-75013 Paris, France
| | | | - Mohamed Doulazmi
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de Biologie Paris Seine, Adaptation Biologique et vieillissement, F-75005 Paris, France
| | - Rosine Wehrlé
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de Biologie Paris Seine, Neuroscience Paris Seine, F-75005 Paris, France
| | - Vidjeacoumary Cannaya
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de Biologie Paris Seine, Neuroscience Paris Seine, F-75005 Paris, France
| | - Corinne Bachelin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, F-75013 Paris, France
| | - Jean-Vianney Barnier
- Institute of Neuroscience Paris-Saclay, CNRS-Université Paris-Sud, UMR9197, F-91405 Orsay, France
| | - Zhengping Jia
- Neurosciences & Mental Health, The Hospital for Sick Children, and Department of Physiology, Faculty of Medicine, University of Toronto, 555 University, Toronto, Ontario M5G 1X8, Canada
| | - Brahim Nait Oumesmar
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, F-75013 Paris, France
| | - Isabelle Dusart
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de Biologie Paris Seine, Neuroscience Paris Seine, F-75005 Paris, France
| | - Lamia Bouslama-Oueghlani
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de Biologie Paris Seine, Neuroscience Paris Seine, F-75005 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, F-75013 Paris, France.
| |
Collapse
|
10
|
Honisch S, Yu W, Liu G, Alesutan I, Towhid ST, Tsapara A, Schleicher S, Handgretinger R, Stournaras C, Lang F. Chorein addiction in VPS13A overexpressing rhabdomyosarcoma cells. Oncotarget 2016; 6:10309-19. [PMID: 25871399 PMCID: PMC4496357 DOI: 10.18632/oncotarget.3582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 02/13/2015] [Indexed: 12/12/2022] Open
Abstract
Chorein encoded by VPS13A (vacuolar protein sorting-associated protein 13A) is defective in chorea-acanthocytosis. Chorein fosters neuronal cell survival, cortical actin polymerization and cell stiffness. In view of its anti-apoptotic effect in neurons, we explored whether chorein is expressed in cancer cells and influences cancer cell survival. RT-PCR was employed to determine transcript levels, specific siRNA to silence chorein, FACS analysis to follow apoptosis and Western blotting to quantify protein abundance. Chorein transcripts were detected in various cancer cell types. The mRNA coding for chorein and chorein protein were most abundant in drug resistant, poorly differentiated human rhabdomyosarcoma cells. Chorein silencing significantly reduced the ratio of phosphorylated (and thus activated) to total phosphoinositide 3 kinase (PI-3K), pointing to inactivation of this crucial pro-survival signaling molecule. Moreover, chorein silencing diminished transcript levels and protein expression of anti-apoptotic BCL-2 and enhanced transcript levels of pro-apoptotic Bax. Silencing of chorein in rhabdomyosarcoma cells was followed by mitochondrial depolarization, caspase 3 activation and stimulation of early and late apoptosis. In conclusion, chorein is expressed in various cancer cells. In cells with high chorein expression levels chorein silencing promotes apoptotic cell death, an effect paralleled by down-regulation of PI-3K activity and BCL-2/Bax expression ratio.
Collapse
Affiliation(s)
- Sabina Honisch
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Willi Yu
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Guilai Liu
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Syeda T Towhid
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Anna Tsapara
- Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Sabine Schleicher
- Department of Hematology and Oncology, Children's Hospital, University Hospital of Tuebingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, Children's Hospital, University Hospital of Tuebingen, Tübingen, Germany
| | - Christos Stournaras
- Department of Physiology, University of Tübingen, Tübingen, Germany.,Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
PAK1 and CtBP1 Regulate the Coupling of Neuronal Activity to Muscle Chromatin and Gene Expression. Mol Cell Biol 2015; 35:4110-20. [PMID: 26416879 DOI: 10.1128/mcb.00354-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/01/2015] [Indexed: 11/20/2022] Open
Abstract
Acetylcholine receptor (AChR) expression in innervated muscle is limited to the synaptic region. Neuron-induced electrical activity participates in this compartmentalization by promoting the repression of AChR expression in the extrasynaptic regions. Here, we show that the corepressor CtBP1 (C-terminal binding protein 1) is present on the myogenin promoter together with repressive histone marks. shRNA-mediated downregulation of CtBP1 expression is sufficient to derepress myogenin and AChR expression in innervated muscle. Upon denervation, CtBP1 is displaced from the myogenin promoter and relocates to the cytoplasm, while repressive histone marks are replaced by activating ones concomitantly to the activation of myogenin expression. We also observed that upon denervation the p21-activated kinase 1 (PAK1) expression is upregulated, suggesting that phosphorylation by PAK1 may be involved in the relocation of CtBP1. Indeed, preventing CtBP1 Ser158 phosphorylation induces CtBP1 accumulation in the nuclei and abrogates the activation of myogenin and AChR expression. Altogether, these findings reveal a molecular mechanism to account for the coordinated control of chromatin modifications and muscle gene expression by presynaptic neurons via a PAK1/CtBP1 pathway.
Collapse
|
12
|
Zhou N, Ding B, Agler M, Cockett M, McPhee F. Lethality of PAK3 and SGK2 shRNAs to human papillomavirus positive cervical cancer cells is independent of PAK3 and SGK2 knockdown. PLoS One 2015; 10:e0117357. [PMID: 25615606 PMCID: PMC4304782 DOI: 10.1371/journal.pone.0117357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/22/2014] [Indexed: 01/23/2023] Open
Abstract
The p21-activated kinase 3 (PAK3) and the serum and glucocorticoid-induced kinase 2 (SGK2) have been previously proposed as essential kinases for human papillomavirus positive (HPV+) cervical cancer cell survival. This was established using a shRNA knockdown approach. To validate PAK3 and SGK2 as potential targets for HPV+ cervical cancer therapy, the relationship between shRNA-induced phenotypes in HPV+ cervical cancer cells and PAK3 or SGK2 knockdown was carefully examined. We observed that the phenotypes of HPV+ cervical cancer cells induced by various PAK3 and SGK2 shRNAs could not be rescued by complement expression of respective cDNA constructs. A knockdown-deficient PAK3 shRNA with a single mismatch was sufficient to inhibit HeLa cell growth to a similar extent as wild-type PAK3 shRNA. The HPV+ cervical cancer cells were also susceptible to several non-human target shRNAs. The discrepancy between PAK3 and SGK2 shRNA-induced apoptosis and gene expression knockdown, as well as cell death stimulation, suggested that these shRNAs killed HeLa cells through different pathways that may not be target-specific. These data demonstrated that HPV+ cervical cancer cell death was not associated with RNAi-induced PAK3 and SGK2 knockdown but likely through off-target effects.
Collapse
Affiliation(s)
- Nannan Zhou
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Bo Ding
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Michele Agler
- Department of Leads Discovery, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Mark Cockett
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Fiona McPhee
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
13
|
Field J, Manser E. The PAKs come of age: Celebrating 18 years of discovery. CELLULAR LOGISTICS 2014; 2:54-58. [PMID: 23125949 PMCID: PMC3485743 DOI: 10.4161/cl.22084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein kinases are versatile signaling molecules that are involved in the regulation most physiological responses. The p21-activated kinases (PAKs) can be activated directly by the small GTPases Rac and Cdc42 and are among the best characterized downstream effectors of these Rho proteins. The structure, substrate specificity and functional role of PAKS are evolutionarily conserved from protozoa to mammals. Vertebrate PAKs are particularly important for cytoskeletal remodeling and focal adhesion assembly, thereby contributing to dynamic processes such as cell migration and synaptic plasticity. This issue of Cellular Logistics focuses on the PAK family of kinases, with ten reviews written by researchers currently working in the field. Here in this introductory overview we highlight some of the most interesting recent discoveries regarding PAK biochemistry and biology. The reviews in this issue cover a range of topics including the atomic structures of PAK1 and PAK4, their role in animals as assessed by knockout studies, and how PAKs are likely to contribute to cancer and neurodegenerative diseases. The promise remains that PAK inhibitors will emerge that validate current pre-clinical studies suggesting that blocking PAK activity will positively contribute to human health.
Collapse
Affiliation(s)
- Jeffrey Field
- Department of Pharmacology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | | |
Collapse
|
14
|
Ribeiro SA, D'Ambrosio MV, Vale RD. Induction of focal adhesions and motility in Drosophila S2 cells. Mol Biol Cell 2014; 25:3861-9. [PMID: 25273555 PMCID: PMC4244196 DOI: 10.1091/mbc.e14-04-0863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, normally immotile S2 cells are engineered to induce the formation of focal adhesions and cell motility by the transfection of a single gene encoding an integrin subunit. It is demonstrated that the focal adhesions recruit expected components and exhibit mechanosensitive behavior on integrin-ligand substrates of different stiffnesses. Focal adhesions are dynamic structures that interact with the extracellular matrix on the cell exterior and actin filaments on the cell interior, enabling cells to adhere and crawl along surfaces. We describe a system for inducing the formation of focal adhesions in normally non–ECM-adherent, nonmotile Drosophila S2 cells. These focal adhesions contain the expected molecular markers such as talin, vinculin, and p130Cas, and they require talin for their formation. The S2 cells with induced focal adhesions also display a nonpolarized form of motility on vitronectin-coated substrates. Consistent with findings in mammalian cells, the degree of motility can be tuned by changing the stiffness of the substrate and was increased after the depletion of PAK3, a p21-activated kinase. A subset of nonmotile, nonpolarized cells also exhibited focal adhesions that rapidly assembled and disassembled around the cell perimeter. Such cooperative and dynamic fluctuations of focal adhesions were decreased by RNA interference (RNAi) depletion of myosin II and focal adhesion kinase, suggesting that this behavior requires force and focal adhesion maturation. These results demonstrate that S2 cells, a cell line that is well studied for cytoskeletal dynamics and readily amenable to protein manipulation by RNAi, can be used to study the assembly and dynamics of focal adhesions and mechanosensitive cell motility.
Collapse
Affiliation(s)
- Susana A Ribeiro
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Michael V D'Ambrosio
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Ronald D Vale
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
15
|
Koth AP, Oliveira BR, Parfitt GM, Buonocore JDQ, Barros DM. Participation of group I p21-activated kinases in neuroplasticity. ACTA ACUST UNITED AC 2014; 108:270-7. [PMID: 25174326 DOI: 10.1016/j.jphysparis.2014.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/25/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022]
Abstract
PAKs are a family of serine/threonine protein kinases activated by small GTPases of the Rho family, including Rac and Cdc42, and are categorized into group I (isoforms 1, 2 and 3) and group II (isoforms 4, 5 and 6). PAK1 and PAK3 are critically involved in biological mechanisms associated with neurodevelopment, neuroplasticity and maturation of the nervous system, and changes in their activity have been detected in pathological disorders, such as Alzheimer's disease, Huntington's disease and mental retardation. The group I PAKs have been associated with neurological processes due to their involvement in intracellular mechanisms that result in molecular and cellular morphological alterations that promote cytoskeletal outgrowth, increasing the efficiency of synaptic transmission. Their substrates in these processes include other intracellular signaling molecules, such as Raf, Mek and LIMK, as well as other components of the cytoskeleton, such as MLC and FLNa. In this review, we describe the characteristics of group I PAKs, such as their molecular structure, mechanisms of activation and importance in the neurobiological processes involved in synaptic plasticity.
Collapse
Affiliation(s)
- André P Koth
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Bruno R Oliveira
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Biologia Molecular, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Gustavo M Parfitt
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Juliana de Quadros Buonocore
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| | - Daniela M Barros
- Instituto de Ciências Biológicas (ICB), Programa de Pós-graduação em Ciências Fisiológicas, Fisiologia Animal Comparada, Laboratório de Neurociências, Universidade Federal do Rio Grande (FURG), Av Itália, Km 8, Rio Grande, RS 96203-900, Brazil.
| |
Collapse
|
16
|
Piccand J, Meunier A, Merle C, Jia Z, Barnier JV, Gradwohl G. Pak3 promotes cell cycle exit and differentiation of β-cells in the embryonic pancreas and is necessary to maintain glucose homeostasis in adult mice. Diabetes 2014; 63:203-15. [PMID: 24163148 PMCID: PMC3968432 DOI: 10.2337/db13-0384] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The transcription factor neurogenin3 (Ngn3) triggers islet cell differentiation in the developing pancreas. However, little is known about the molecular mechanisms coupling cell cycle exit and differentiation in Ngn3(+) islet progenitors. We identified a novel effector of Ngn3 endocrinogenic function, the p21 protein-activated kinase Pak3, known to control neuronal differentiation and implicated in X-linked intellectual disability in humans. We show that Pak3 expression is initiated in Ngn3(+) endocrine progenitor cells and next maintained in maturing hormone-expressing cells during pancreas development as well as in adult islet cells. In Pak3-deficient embryos, the proliferation of Ngn3(+) progenitors and β-cells is transiently increased concomitantly with an upregulation of Ccnd1. β-Cell differentiation is impaired at E15.5 but resumes at later stages. Pak3-deficient mice do not develop overt diabetes but are glucose intolerant under high-fat diet (HFD). In the intestine, Pak3 is expressed in enteroendocrine cells but is not necessary for their differentiation. Our results indicate that Pak3 is a novel regulator of β-cell differentiation and function. Pak3 acts downstream of Ngn3 to promote cell cycle exit and differentiation in the embryo by a mechanism that might involve repression of Ccnd1. In the adult, Pak3 is required for the proper control of glucose homeostasis under challenging HFD.
Collapse
Affiliation(s)
- Julie Piccand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
| | - Aline Meunier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
| | - Carole Merle
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
| | - Zhengping Jia
- Neurosciences and Mental Health, The Hospital for Sick Children, Department of Physiology, University of Toronto, Toronto, Canada
| | - Jean-Vianney Barnier
- Université Paris-Sud, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France
- Centre National de Recherche Scientifique, UMR 8195, Orsay, France
| | - Gérard Gradwohl
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
- Corresponding author: Gérard Gradwohl,
| |
Collapse
|
17
|
Role of p-21-activated kinases in cancer progression. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 309:347-87. [PMID: 24529727 DOI: 10.1016/b978-0-12-800255-1.00007-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The p-21-activated kinases (PAKs) are downstream effectors of Rho GTPases Rac and Cdc42. The PAK family consists of six members which are segregated into two subgroups (Group I and Group II) based on sequence homology. Group I PAKs (PAK1-3) are the most extensively studied but there is increasing interest in the functionality of Group II PAKs (PAK4-6). The PAK family proteins are thought to play an important role in many different cellular processes, some of which have particular significance in the context of cancer progression. This review explores established and more recent data, linking the PAK family kinases to cancer progression including expression profiles, evasion of apoptosis, promotion of cell survival, and regulation of cell invasion. Finally, we discuss attempts to therapeutically target the PAK family and outline the major obstacles that still need to be overcome.
Collapse
|
18
|
Shin YJ, Kim YB, Kim JH. Protein kinase CK2 phosphorylates and activates p21-activated kinase 1. Mol Biol Cell 2013; 24:2990-9. [PMID: 23885116 PMCID: PMC3771959 DOI: 10.1091/mbc.e13-04-0204] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Activation of the p21-activated kinase 1 (PAK1) is achieved through a conformational change that converts an inactive PAK1 dimer to an active monomer. In this paper, we show that this change is necessary but not sufficient to activate PAK1 and that it is, rather, required for CK2-dependent PAK1(S223) phosphorylation that converts a monomeric PAK1 into a catalytically active form. This phosphorylation appears to be essential for autophosphorylation at specific residues and overall activity of PAK1. A phosphomimetic mutation (S223E) bypasses the requirement for GTPases in PAK1 activation, whereas the constitutive activity of the PAK1 mutant (PAK1(H83,86L)), postulated to mimic GTPase-induced structural changes, is abolished by inhibition of S223 phosphorylation. Thus, S223 is likely accessible to CK2 upon conformational changes of PAK1 induced by GTPase-dependent and GTPase-independent stimuli, suggesting that S223 phosphorylation may play a key role in the final step of the PAK1 activation process. The physiological significance of this phosphorylation is reinforced by the observations that CK2 is responsible for epidermal growth factor-induced PAK1 activation and that inhibition of S223 phosphorylation abrogates PAK1-mediated malignant transformation of prostate epithelial cells. Taken together, these findings identify CK2 as an upstream activating kinase of PAK1, providing a novel mechanism for PAK1 activation.
Collapse
Affiliation(s)
- Yong Jae Shin
- Department of Biochemistry and Molecular Medicine, George Washington University Medical Center, Washington, DC 20037
| | | | | |
Collapse
|
19
|
Schmidt EM, Schmid E, Münzer P, Hermann A, Eyrich AK, Russo A, Walker B, Gu S, vom Hagen JM, Faggio C, Schaller M, Föller M, Schöls L, Gawaz M, Borst O, Storch A, Stournaras C, Lang F. Chorein sensitivity of cytoskeletal organization and degranulation of platelets. FASEB J 2013; 27:2799-806. [PMID: 23568775 DOI: 10.1096/fj.13-229286] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Chorea-acanthocytosis (ChAc), a lethal disease caused by defective chorein, is characterized by neurodegeneration and erythrocyte acanthocytosis. The functional significance of chorein in other cell types remained ill-defined. The present study revealed chorein expression in blood platelets. As compared to platelets from healthy volunteers, platelets from patients with ChAc displayed a 47% increased globular/filamentous actin ratio, indicating actin depolymerization. Moreover, phosphoinositide-3-kinase subunit p85 phosphorylation, p21 protein-activated kinase (PAK1) phosphorylation, as well as vesicle-associated membrane protein 8 (VAMP8) expression were significantly reduced in platelets from patients with ChAc (by 17, 22, and 39%, respectively) and in megakaryocytic (MEG-01) cells following chorein silencing (by 16, 54, and 11%, respectively). Activation-induced platelet secretion from dense granules (ATP release) and α granules (P-selectin exposure) were significantly less (by 55% after stimulation with 1 μg/ml CRP and by 33% after stimulation with 5 μM TRAP, respectively) in ChAc platelets than in control platelets. Furthermore, platelet aggregation following stimulation with different platelet agonists was significantly impaired. These observations reveal a completely novel function of chorein, i.e., regulation of secretion and aggregation of blood platelets.
Collapse
Affiliation(s)
- Eva-Maria Schmidt
- Department of Physiology, University of Tübingen, Gmelinstrasse 5, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kelemen O, Convertini P, Zhang Z, Wen Y, Shen M, Falaleeva M, Stamm S. Function of alternative splicing. Gene 2013; 514:1-30. [PMID: 22909801 PMCID: PMC5632952 DOI: 10.1016/j.gene.2012.07.083] [Citation(s) in RCA: 524] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/21/2012] [Accepted: 07/30/2012] [Indexed: 12/15/2022]
Abstract
Almost all polymerase II transcripts undergo alternative pre-mRNA splicing. Here, we review the functions of alternative splicing events that have been experimentally determined. The overall function of alternative splicing is to increase the diversity of mRNAs expressed from the genome. Alternative splicing changes proteins encoded by mRNAs, which has profound functional effects. Experimental analysis of these protein isoforms showed that alternative splicing regulates binding between proteins, between proteins and nucleic acids as well as between proteins and membranes. Alternative splicing regulates the localization of proteins, their enzymatic properties and their interaction with ligands. In most cases, changes caused by individual splicing isoforms are small. However, cells typically coordinate numerous changes in 'splicing programs', which can have strong effects on cell proliferation, cell survival and properties of the nervous system. Due to its widespread usage and molecular versatility, alternative splicing emerges as a central element in gene regulation that interferes with almost every biological function analyzed.
Collapse
Affiliation(s)
- Olga Kelemen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Paolo Convertini
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Zhaiyi Zhang
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Yuan Wen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Manli Shen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Marina Falaleeva
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Stefan Stamm
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
21
|
Combeau G, Kreis P, Domenichini F, Amar M, Fossier P, Rousseau V, Barnier JV. The p21-activated kinase PAK3 forms heterodimers with PAK1 in brain implementing trans-regulation of PAK3 activity. J Biol Chem 2012; 287:30084-96. [PMID: 22815483 DOI: 10.1074/jbc.m112.355073] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
p21-activated kinase 1 (PAK1) and PAK3 belong to group I of the PAK family and control cell movement and division. They also regulate dendritic spine formation and maturation in the brain, and play a role in synaptic transmission and synaptic plasticity. PAK3, in particular, is known for its implication in X-linked intellectual disability. The pak3 gene is expressed in neurons as a GTPase-regulated PAK3a protein and also as three splice variants which display constitutive kinase activity. PAK1 regulation is based on its homodimerization, forming an inactive complex. Here, we analyze the PAK3 capacity to dimerize and show that although PAK3a is able to homodimerize, it is more likely to form heterodimeric complexes with PAK1. We further show that two intellectual disability mutations impair dimerization with PAK1. The b and c inserts present in the regulatory domain of PAK3 splice variants decrease the dimerization but retain the capacity to form heterodimers with PAK1. PAK1 and PAK3 are co-expressed in neurons, are colocalized within dendritic spines, co-purify with post-synaptic densities, and co-immunoprecipitate in brain lysates. Using kinase assays, we demonstrate that PAK1 inhibits the activity of PAK3a but not of the splice variant PAK3b in a trans-regulatory manner. Altogether, these results show that PAK3 and PAK1 signaling may be coordinated by heterodimerization.
Collapse
Affiliation(s)
- Gaëlle Combeau
- Centre de Neurosciences Paris-Sud, Université Paris-Sud, UMR 8195 and CNRS, UMR 8195, Orsay F-91405, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Föller M, Hermann A, Gu S, Alesutan I, Qadri SM, Borst O, Schmidt E, Schiele F, Hagen JMV, Saft C, Schöls L, Lerche H, Stournaras C, Storch A, Lang F. Chorein‐sensitive polymerization of cortical actin and suicidal cell death in chorea‐acanthocytosis. FASEB J 2012; 26:1526-34. [DOI: 10.1096/fj.11-198317] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michael Föller
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Andreas Hermann
- Division of Neurodegenerative DiseasesDepartment of NeurologyUniversity of TechnologyDresdenGermany
| | - Shuchen Gu
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Ioana Alesutan
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Syed M. Qadri
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Oliver Borst
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | | | - Franziska Schiele
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Jennifer Müller vom Hagen
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Carsten Saft
- Department of NeurologyRuhr UniversityBochumGermany
| | - Ludger Schöls
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- DZNEResearch Site TübingenTübingenGermany
| | - Holger Lerche
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | | | - Alexander Storch
- Division of Neurodegenerative DiseasesDepartment of NeurologyUniversity of TechnologyDresdenGermany
- German Center for Neurodegenerative Diseases (DZNE)Research Site DresdenDresdenGermany
| | - Florian Lang
- Department of PhysiologyUniversity of TübingenTübingenGermany
| |
Collapse
|
23
|
Thévenot E, Moreau AW, Rousseau V, Combeau G, Domenichini F, Jacquet C, Goupille O, Amar M, Kreis P, Fossier P, Barnier JV. p21-Activated kinase 3 (PAK3) protein regulates synaptic transmission through its interaction with the Nck2/Grb4 protein adaptor. J Biol Chem 2011; 286:40044-59. [PMID: 21949127 DOI: 10.1074/jbc.m111.262246] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations in the p21-activated kinase 3 gene (pak3) are responsible for nonsyndromic forms of mental retardation. Expression of mutated PAK3 proteins in hippocampal neurons induces abnormal dendritic spine morphology and long term potentiation anomalies, whereas pak3 gene invalidation leads to cognitive impairments. How PAK3 regulates synaptic plasticity is still largely unknown. To better understand how PAK3 affects neuronal synaptic plasticity, we focused on its interaction with the Nck adaptors that play a crucial role in PAK signaling. We report here that PAK3 interacts preferentially with Nck2/Grb4 in brain extracts and in transfected cells. This interaction is independent of PAK3 kinase activity. Selective uncoupling of the Nck2 interactions in acute cortical slices using an interfering peptide leads to a rapid increase in evoked transmission to pyramidal neurons. The P12A mutation in the PAK3 protein strongly decreases the interaction with Nck2 but only slightly with Nck1. In transfected hippocampal cultures, expression of the P12A-mutated protein has no effect on spine morphogenesis or synaptic density. The PAK3-P12A mutant does not affect synaptic transmission, whereas the expression of the wild-type PAK3 protein decreases the amplitude of spontaneous miniature excitatory currents. Altogether, these data show that PAK3 down-regulates synaptic transmission through its interaction with Nck2.
Collapse
Affiliation(s)
- Emmanuel Thévenot
- CNRS, Institut de Neurobiologie Alfred Fessard, Laboratoire de Neurobiologie Cellulaire et Moléculaire, 91190 Gif sur Yvette, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Dasouki MJ, Lushington GH, Hovanes K, Casey J, Gorre M. The 3q29 microdeletion syndrome: report of three new unrelated patients and in silico "RNA binding" analysis of the 3q29 region. Am J Med Genet A 2011; 155A:1654-60. [PMID: 21626679 DOI: 10.1002/ajmg.a.34080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2009] [Accepted: 04/04/2011] [Indexed: 12/17/2022]
Abstract
The human 3q29 microdeletion syndrome is associated with mild facial dysmorphism, developmental delay and variable congenital malformations. We report three new unrelated patients with this syndrome. We also performed in silico RNA binding analysis in silico on the 3q29 critical region genes. Several genes within this genomic region including DLG1 and RNF168 are predicted to bind RNA. While recessive mutations in RNF168 cause RIDDLE syndrome, an immune deficiency and radiosensitivity disorder, the potential impact of heterozygous deletion of RNF168 on patients with the 3q29 deletion syndrome is still unknown.
Collapse
Affiliation(s)
- Majed J Dasouki
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, USA.
| | | | | | | | | |
Collapse
|
25
|
Li T, Zhang J, Zhu F, Wen W, Zykova T, Li X, Liu K, Peng C, Ma W, Shi G, Dong Z, Bode AM, Dong Z. P21-activated protein kinase (PAK2)-mediated c-Jun phosphorylation at 5 threonine sites promotes cell transformation. Carcinogenesis 2010; 32:659-66. [PMID: 21177766 DOI: 10.1093/carcin/bgq271] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The oncoprotein c-Jun is one of the components of the activator protein-1 (AP-1) transcription factor complex. AP-1 regulates the expression of many genes and is involved in a variety of biological functions such as cell transformation, proliferation, differentiation and apoptosis. AP-1 activates a variety of tumor-related genes and therefore promotes tumorigenesis and malignant transformation. Here, we found that epidermal growth factor (EGF) induces phosphorylation of c-Jun by P21-activated kinase (PAK) 2. Our data showed that PAK2 binds and phosphorylates c-Jun at five threonine sites (Thr2, Thr8, Thr89, Thr93 and Thr286) in vitro and ex vivo. Knockdown of PAK2 in JB6 Cl41 (P+) cells had no effect on c-Jun phosphorylation at Ser63 or Ser73 but resulted in decreases in EGF-induced anchorage-independent cell transformation, proliferation and AP-1 activity. Mutation at all five c-Jun threonine sites phosphorylated by PAK2 decreased the transforming ability of JB6 cells. Knockdown of PAK2 in SK-MEL-5 melanoma cells also decreased colony formation, proliferation and AP-1 activity. These results indicated that PAK2/c-Jun signaling plays an important role in EGF-induced cell proliferation and transformation.
Collapse
Affiliation(s)
- Tingting Li
- The Hormel Institute, University of Minnesota, 801 16th Avenue North East, Austin, MN 55912, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kong L, Lovell PV, Heger A, Mello CV, Ponting CP. Accelerated evolution of PAK3- and PIM1-like kinase gene families in the zebra finch, Taeniopygia guttata. Mol Biol Evol 2010; 27:1923-34. [PMID: 20237222 DOI: 10.1093/molbev/msq080] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Genes encoding protein kinases tend to evolve slowly over evolutionary time, and only rarely do they appear as recent duplications in sequenced vertebrate genomes. Consequently, it was a surprise to find two families of kinase genes that have greatly and recently expanded in the zebra finch (Taeniopygia guttata) lineage. In contrast to other amniotic genomes (including chicken) that harbor only single copies of p21-activated serine/threonine kinase 3 (PAK3) and proviral integration site 1 (PIM1) genes, the zebra finch genome appeared at first to additionally contain 67 PAK3-like (PAK3L) and 51 PIM1-like (PIM1L) protein kinase genes. An exhaustive analysis of these gene models, however, revealed most to be incomplete, owing to the absence of terminal exons. After reprediction, 31 PAK3L genes and 10 PIM1L genes remain, and all but three are predicted, from the retention of functional sites and open reading frames, to be enzymatically active. PAK3L, but not PIM1L, gene sequences show evidence of recurrent episodes of positive selection, concentrated within structures spatially adjacent to N- and C-terminal protein regions that have been discarded from zebra finch PAK3L genes. At least seven zebra finch PAK3L genes were observed to be expressed in testis, whereas two sequences were found transcribed in the brain, one broadly including the song nuclei and the other in the ventricular zone and in cells resembling Bergmann's glia in the cerebellar Purkinje cell layer. Two PIM1L sequences were also observed to be expressed with broad distributions in the zebra finch brain, one in both the ventricular zone and the cerebellum and apparently associated with glial cells and the other showing neuronal cell expression and marked enrichment in midbrain/thalamic nuclei. These expression patterns do not correlate with zebra finch-specific features such as vocal learning. Nevertheless, our results show how ancient and conserved intracellular signaling molecules can be co-opted, following duplication, thereby resulting in lineage-specific functions, presumably affecting the zebra finch testis and brain.
Collapse
Affiliation(s)
- Lesheng Kong
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | | | | |
Collapse
|
27
|
Anamika K, Garnier N, Srinivasan N. Functional diversity of human protein kinase splice variants marks significant expansion of human kinome. BMC Genomics 2009; 10:622. [PMID: 20028505 PMCID: PMC2805699 DOI: 10.1186/1471-2164-10-622] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 12/22/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Protein kinases are involved in diverse spectrum of cellular processes. Availability of draft version of the human genomic data in the year 2001 enabled recognition of repertoire of protein kinases. However, over the years the human genomic data is being refined and the current release of human genomic data has helped us to recognize a larger repertoire of over 900 human protein kinases represented mainly by splice variants. RESULTS Many of these identified protein kinases are alternatively spliced products. Interestingly, some of the human kinase splice variants appear to be significantly diverged in terms of their functional properties as represented by incorporation or absence of one or more domains. Many sets of protein kinase splice variants have substantially different domain organization and in a few sets of splice variants kinase domains belong to different subfamilies of kinases suggesting potential participation in different signal transduction pathways. CONCLUSIONS Addition or deletion of a domain between splice variants of multi-domain kinases appears to be a means of generating differences in the functional features of otherwise similar kinases. It is intriguing that marked sequence diversity within the catalytic regions of some of the splice variant kinases result in kinases belonging to different subfamilies. These human kinase splice variants with different functions might contribute to diversity of eukaryotic cellular signaling.
Collapse
|
28
|
Demyanenko GP, Halberstadt AI, Rao RS, Maness PF. CHL1 cooperates with PAK1-3 to regulate morphological differentiation of embryonic cortical neurons. Neuroscience 2009; 165:107-15. [PMID: 19819308 DOI: 10.1016/j.neuroscience.2009.09.077] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 09/04/2009] [Accepted: 09/27/2009] [Indexed: 01/27/2023]
Abstract
The cell adhesion molecule close homologue of L1 (CHL1) is important for apical dendritic projection and laminar positioning of pyramidal neurons in caudal regions of the cerebral cortex. The p21-activated kinase (PAK1-3) subfamily of serine/threonine kinases has also been implicated in regulating cell adhesion, migration, and morphology. Immunofluorescence staining in mouse embryonic brain showed that PAK1-3 was expressed in embryonic cortex and colocalized with CHL1 during neuronal migration and differentiation. To investigate a cooperative function for CHL1 and PAK in pyramidal cell differentiation or migration, a dominant-negative PAK mutant (PAK1 AID) that inhibits PAK1-3 kinase activity while coexpressing a green fluorescent protein (GFP) reporter was electroporated into the lateral ventricles of wild type (WT) and CHL1 null mutant mouse embryos (E14.5), then brain slices were cultured and neurons analyzed for laminar positioning and morphology by confocal microscopy after 3 days in vitro. Expression of PAK1 AID in CHL1 mutant cortex inactivated PAK and caused embryonic cortical neurons to branch profusely in the intermediate zone (IZ) and cortical plate (CP). The number of nodes, terminals and length of leading processes/apical dendrites of CHL1 mutant embryos expressing PAK1 AID increased dramatically, compared to CHL1 mutants without PAK1 AID, or WT embryos with or without PAK1 AID. These findings suggest that CHL1 and PAK1-3 kinase cooperate, most likely in independent pathways, in regulating morphological development of the leading process/apical dendrite of embryonic cortical neurons.
Collapse
Affiliation(s)
- G P Demyanenko
- Department of Biochemistry and Biophysics, The University of North Carolina School of Medicine at Chapel Hill, 27599, USA
| | | | | | | |
Collapse
|
29
|
Abstract
The p21-activated kinase (PAK) family of serine/threonine kinases is important in physiological processes including motility, survival, mitosis, transcription and translation. PAKs are evolutionally conserved and widely expressed in a variety of tissues and are often overexpressed in multiple cancer types. Depending on structural and functional similarities, the six members of PAK family are divided into two groups with three members in each group. Group I PAKs are activated by extracellular signals through GTPase-dependent and GTPase-independent mechanisms. In contrast, group II PAKs are constitutively active. Over the years, accumulating data from tissue culture models and human tumors has increased our understanding about the biology of PAK family members. In this review, we have summarized the complex regulation of PAK and its downstream diverse myriads of effectors, which in turn are responsible for the biological effects of PAK family of kinases in cancer cells.
Collapse
|
30
|
Tissue and developmental regulation of fragile X mental retardation 1 exon 12 and 15 isoforms. Neurobiol Dis 2009; 35:52-62. [PMID: 19362146 DOI: 10.1016/j.nbd.2009.03.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 03/24/2009] [Accepted: 03/29/2009] [Indexed: 11/22/2022] Open
Abstract
The pre-mRNA of the fragile X mental retardation 1 gene (FMR1) is subject to exon skipping and alternative splice site selection, which can generate up to 12 isoforms. The expression and function of these variants in vivo has not yet been fully explored. In the present study, we investigated the distribution of Fmr1 exon 12 and exon 15 isoforms. Exon 12 encodes an extension of KH(2) domain, one of the RNA binding domains in the FMR1 gene product (FMRP) and we show that exon 12 variant proteins differentially interact with kissing complex RNA. Alternative splicing at exon 15 produces FMRPs differing in RNA binding ability and each is distinguished by unique post-translational modifications. Using semiquantitative RT-PCR and Northern blotting, we found that particular Fmr1 exon 12 and exon 15 isoforms change during neuronal differentiation. Interestingly, Fmr1 exon 12 variants display tissue-specific and developmental differences, while exon 15-containing transcripts vary less. Altogether, the spatio-temporal plasticity of FMR1 mRNA is consistent with complex RNA processing that is mis-regulated in fragile X syndrome.
Collapse
|
31
|
Humeau Y, Gambino F, Chelly J, Vitale N. X-linked mental retardation: focus on synaptic function and plasticity. J Neurochem 2009; 109:1-14. [DOI: 10.1111/j.1471-4159.2009.05881.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Kreis P, Barnier JV. PAK signalling in neuronal physiology. Cell Signal 2008; 21:384-93. [PMID: 19036346 DOI: 10.1016/j.cellsig.2008.11.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Accepted: 11/06/2008] [Indexed: 12/11/2022]
Abstract
Group I p21-activated kinases are a family of key effectors of Rac1 and Cdc42 and they regulate many aspects of cellular function, such as cytoskeleton dynamics, cell movement and cell migration, cell proliferation and differentiation, and gene expression. The three genes PAK1/2/3 are expressed in brain and recent evidence indicates their crucial roles in neuronal cell fate, in axonal guidance and neuronal polarisation, and in neuronal migration. Moreover they are implicated in neurodegenerative diseases and play an important role in synaptic plasticity, with PAK3 being specifically involved in mental retardation. The main goal of this review is to describe the molecular mechanisms that govern the different functions of group I PAK in neuronal signalling and to discuss the specific functions of each isoform.
Collapse
Affiliation(s)
- Patricia Kreis
- CNRS, Institut de Neurobiologie Alfred Fessard-FRC2118, Laboratoire de Neurobiologie Cellulaire et Moléculaire-UPR9040, Gif sur Yvette, France.
| | | |
Collapse
|