1
|
Blanco FA, Saifullah MAB, Cheng JX, Abella C, Scala F, Firozi K, Niu S, Park J, Chin J, Tolias KF. Targeting Tiam1 Enhances Hippocampal-Dependent Learning and Memory in the Adult Brain and Promotes NMDA Receptor-Mediated Synaptic Plasticity and Function. J Neurosci 2025; 45:e0298242024. [PMID: 39725519 PMCID: PMC11800756 DOI: 10.1523/jneurosci.0298-24.2024] [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: 02/14/2024] [Revised: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Excitatory synapses and the actin-rich dendritic spines on which they reside are indispensable for information processing and storage in the brain. In the adult hippocampus, excitatory synapses must balance plasticity and stability to support learning and memory. However, the mechanisms governing this balance remain poorly understood. Tiam1 is an actin cytoskeleton regulator prominently expressed in the dentate gyrus (DG) throughout life. Previously, we showed that Tiam1 promotes dentate granule cell synapse and spine stabilization during development, but its role in the adult hippocampus remains unclear. Here, we deleted Tiam1 from adult forebrain excitatory neurons (Tiam1fKO ) and assessed the effects on hippocampal-dependent behaviors. Adult male and female Tiam1fKO mice displayed enhanced contextual fear memory, fear extinction, and spatial discrimination. Investigation into underlying mechanisms revealed that dentate granule cells from Tiam1fKO brain slices exhibited augmented synaptic plasticity and N-methyl-D-aspartate-type glutamate receptor (NMDAR) function. Additionally, Tiam1 loss in primary hippocampal neurons blocked agonist-induced NMDAR internalization, reduced filamentous actin levels, and promoted activity-dependent spine remodeling. Notably, strong NMDAR activation in wild-type hippocampal neurons triggered Tiam1 loss from spines. Our results suggest that Tiam1 normally constrains hippocampal-dependent learning and memory in the adult brain by restricting NMDAR-mediated synaptic plasticity in the DG. We propose that Tiam1 achieves this by limiting NMDAR availability at synaptic membranes and stabilizing spine actin cytoskeleton and that these constraints can be alleviated by activity-dependent degradation of Tiam1. These findings reveal a previously unknown mechanism restricting hippocampal synaptic plasticity and highlight Tiam1 as a therapeutic target for enhancing cognitive function.
Collapse
Affiliation(s)
- Francisco A Blanco
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | | | - Jinxuan X Cheng
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Carlota Abella
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Federico Scala
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Sanyong Niu
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Jin Park
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Memory & Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Memory & Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
2
|
Baudouin L, Adès N, Kanté K, Bachelin C, Hmidan H, Deboux C, Panic R, Ben Messaoud R, Velut Y, Hamada S, Pionneau C, Duarte K, Poëa-Guyon S, Barnier JV, Nait Oumesmar B, Bouslama-Oueghlani L. Antagonistic actions of PAK1 and NF2/Merlin drive myelin membrane expansion in oligodendrocytes. Glia 2024; 72:1518-1540. [PMID: 38794866 DOI: 10.1002/glia.24570] [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: 01/07/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lucas Baudouin
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Noémie Adès
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Kadia Kanté
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Corinne Bachelin
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Hatem Hmidan
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
- Al-Quds University, Faculty of Medicine, Jerusalem, Palestine
| | - Cyrille Deboux
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Radmila Panic
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Rémy Ben Messaoud
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Yoan Velut
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Soumia Hamada
- Sorbonne Université, Inserm, UMS Production et Analyse des Données en Sciences de la vie et en Santé, PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, Paris, France
| | - Cédric Pionneau
- Sorbonne Université, Inserm, UMS Production et Analyse des Données en Sciences de la vie et en Santé, PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, Paris, France
| | - Kévin Duarte
- 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
| | - Jean-Vianney Barnier
- Institut des Neurosciences Paris-Saclay, UMR 9197, CNRS, Université Paris-Saclay, Saclay, France
| | - Brahim Nait Oumesmar
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lamia Bouslama-Oueghlani
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
3
|
Abstract
Neuropathic pain is a debilitating form of pain arising from injury or disease of the nervous system that affects millions of people worldwide. Despite its prevalence, the underlying mechanisms of neuropathic pain are still not fully understood. Dendritic spines are small protrusions on the surface of neurons that play an important role in synaptic transmission. Recent studies have shown that dendritic spines reorganize in the superficial and deeper laminae of the spinal cord dorsal horn with the development of neuropathic pain in multiple models of disease or injury. Given the importance of dendritic spines in synaptic transmission, it is possible that studying dendritic spines could lead to new therapeutic approaches for managing intractable pain. In this review article, we highlight the emergent role of dendritic spines in neuropathic pain, as well as discuss the potential for studying dendritic spines for the development of new therapeutics.
Collapse
Affiliation(s)
- Curtis A Benson
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Jared F King
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Marike L Reimer
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Sierra D Kauer
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Andrew M Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
4
|
Wang Y, Kim B, Gong S, Park J, Zhu M, Wong EM, Park AY, Chernoff J, Guo F. Control of OPC proliferation and repopulation by the intellectual disability gene PAK1 under homeostatic and demyelinating conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591153. [PMID: 38746444 PMCID: PMC11092442 DOI: 10.1101/2024.04.26.591153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Appropriate proliferation and repopulation of oligodendrocyte progenitor cells (OPCs) determine successful (re)myelination in homeostatic and demyelinating brains. Activating mutations in p21-activated kinase 1 (PAK1) cause intellectual disability, neurodevelopmental abnormality, and white matter anomaly in children. It remains unclear if and how PAK1 regulates oligodendroglial development. Here, we report that PAK1 controls proliferation and regeneration of OPCs. Unlike differentiating oligodendrocytes, OPCs display high PAK1 activity which maintains them in a proliferative state by modulating PDGFRa-mediated mitogenic signaling. PAK1-deficient or kinase-inhibited OPCs reduce their proliferation capacity and population expansion. Mice carrying OPC-specific PAK1 deletion or kinase inhibition are populated with fewer OPCs in the homeostatic and demyelinated CNS than control mice. Together, our findings suggest that kinase-activating PAK1 mutations stall OPCs in a progenitor state, impacting timely oligodendroglial differentiation in the CNS of affected children and that PAK1 is a potential molecular target for replenishing OPCs in demyelinating lesions.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Bokyung Kim
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Shuaishuai Gong
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Joohyun Park
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Meina Zhu
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Evelyn M. Wong
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Audrey Y. Park
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| | - Jonathan Chernoff
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111
| | - Fuzheng Guo
- Department of Neurology, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, CA 95817
| |
Collapse
|
5
|
King CP, Chitre AS, Leal-Gutiérrez JD, Tripi JA, Hughson AR, Horvath AP, Lamparelli AC, George A, Martin C, Pierre CLS, Sanches T, Bimschleger HV, Gao J, Cheng R, Nguyen KM, Holl KL, Polesskaya O, Ishiwari K, Chen H, Woods LCS, Palmer AA, Robinson TE, Flagel SB, Meyer PJ. Genomic Loci Influencing Cue-Reactivity in Heterogeneous Stock Rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584852. [PMID: 38559127 PMCID: PMC10980002 DOI: 10.1101/2024.03.13.584852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Addiction vulnerability is associated with the tendency to attribute incentive salience to reward predictive cues; both addiction and the attribution of incentive salience are influenced by environmental and genetic factors. To characterize the genetic contributions to incentive salience attribution, we performed a genome-wide association study (GWAS) in a cohort of 1,645 genetically diverse heterogeneous stock (HS) rats. We tested HS rats in a Pavlovian conditioned approach task, in which we characterized the individual responses to food-associated stimuli ("cues"). Rats exhibited either cue-directed "sign-tracking" behavior or food-cup directed "goal-tracking" behavior. We then used the conditioned reinforcement procedure to determine whether rats would perform a novel operant response for unrewarded presentations of the cue. We found that these measures were moderately heritable (SNP heritability, h2 = .189-.215). GWAS identified 14 quantitative trait loci (QTLs) for 11 of the 12 traits we examined. Interval sizes of these QTLs varied widely. 7 traits shared a QTL on chromosome 1 that contained a few genes (e.g. Tenm4, Mir708) that have been associated with substance use disorders and other mental health traits in humans. Other candidate genes (e.g. Wnt11, Pak1) in this region had coding variants and expression-QTLs in mesocorticolimbic regions of the brain. We also conducted a Phenome-Wide Association Study (PheWAS) on other behavioral measures in HS rats and found that regions containing QTLs on chromosome 1 were also associated with nicotine self-administration in a separate cohort of HS rats. These results provide a starting point for the molecular genetic dissection of incentive salience and provide further support for a relationship between attribution of incentive salience and drug abuse-related traits.
Collapse
Affiliation(s)
- Christopher P. King
- Department of Psychology, University at Buffalo, Buffalo, USA
- Clinical and Research Institute on Addictions, Buffalo, USA
| | - Apurva S. Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | | | - Jordan A. Tripi
- Department of Psychology, University at Buffalo, Buffalo, USA
| | - Alesa R. Hughson
- Department of Psychology, University of Michigan, Ann Arbor, USA
| | - Aidan P. Horvath
- Department of Psychology, University of Michigan, Ann Arbor, USA
| | | | - Anthony George
- Clinical and Research Institute on Addictions, Buffalo, USA
| | - Connor Martin
- Clinical and Research Institute on Addictions, Buffalo, USA
| | | | - Thiago Sanches
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | | | - Jianjun Gao
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | - Katie L. Holl
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, USA
| | - Keita Ishiwari
- Clinical and Research Institute on Addictions, Buffalo, USA
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo USA
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Science Center, Memphis, USA
| | - Leah C. Solberg Woods
- Department of Internal Medicine, Molecular Medicine, Center on Diabetes, Obesity and Metabolism, Wake Forest School of Medicine, Winston-Salem, USA
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, USA
| | | | - Shelly B. Flagel
- Department of Psychiatry, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
| | - Paul J. Meyer
- Department of Psychology, University at Buffalo, Buffalo, USA
| |
Collapse
|
6
|
Cyclin Y regulates spatial learning and memory flexibility through distinct control of the actin pathway. Mol Psychiatry 2023; 28:1351-1364. [PMID: 36434054 PMCID: PMC10005959 DOI: 10.1038/s41380-022-01877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022]
Abstract
Spatial learning and memory flexibility are known to require long-term potentiation (LTP) and long-term depression (LTD), respectively, on a cellular basis. We previously showed that cyclin Y (CCNY), a synapse-remodeling cyclin, is a novel actin-binding protein and an inhibitory regulator of functional and structural LTP in vitro. In this study, we report that Ccny knockout (KO) mice exhibit enhanced LTP and weak LTD at Schaffer collateral-CA1 synapses in the hippocampus. In accordance with enhanced LTP, Ccny KO mice showed improved spatial learning and memory. However, although previous studies reported that normal LTD is necessary for memory flexibility, Ccny KO mice intriguingly showed improved memory flexibility, suggesting that weak LTD could exert memory flexibility when combined with enhanced LTP. At the molecular level, CCNY modulated spatial learning and memory flexibility by distinctively affecting the cofilin-actin signaling pathway in the hippocampus. Specifically, CCNY inhibited cofilin activation by original learning, but reversed such inhibition by reversal learning. Furthermore, viral-mediated overexpression of a phosphomimetic cofilin-S3E in hippocampal CA1 regions enhanced LTP, weakened LTD, and improved spatial learning and memory flexibility, thus mirroring the phenotype of Ccny KO mice. In contrast, the overexpression of a non-phosphorylatable cofilin-S3A in hippocampal CA1 regions of Ccny KO mice reversed the synaptic plasticity, spatial learning, and memory flexibility phenotypes observed in Ccny KO mice. Altogether, our findings demonstrate that LTP and LTD cooperatively regulate memory flexibility. Moreover, CCNY suppresses LTP while facilitating LTD in the hippocampus and negatively regulates spatial learning and memory flexibility through the control of cofilin-actin signaling, proposing CCNY as a learning regulator modulating both memorizing and forgetting processes.
Collapse
|
7
|
Multi-proteomic Analysis Revealed Distinct Protein Profiles in Cerebrospinal Fluid of Patients Between Anti-NMDAR Encephalitis NORSE and Cryptogenic NORSE. Mol Neurobiol 2023; 60:98-115. [PMID: 36224320 DOI: 10.1007/s12035-022-03011-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/21/2022] [Indexed: 12/30/2022]
Abstract
New-onset refractory status epilepticus (NORSE) is rare but intractable. Anti-N-methyl-d-aspartate receptor (NMDAR) encephalitis and cryptogenic etiologies are the two major causes of NORSE with distinct clinical features. To elucidate the underlying mechanisms, 6 patients with anti-NMDAR encephalitis NORSE and 5 with cryptogenic NORSE (C-NORSE) were enrolled. Five patients of cerebrovascular disorders were used as controls. Quantitative proteomic analysis of the cerebrospinal fluid (CSF) samples of the patients revealed 101 and 56 proteins were changed, respectively. The average fold-change of the upregulated proteins, namely up-proteomic score in this study, was positively correlated with the severity and prognosis of the diseases, including ICU stay (r = 0.9308, P = 0.0035 in NMDAR group; r = 0.8977, P = 0.0193 in C-NORSE group), mRS score at discharge (r = 0.9710, P = 0.0111 in NMDAR group; r = 0.7071, P = 0.2000 in C-NORSE group), and time taken for patients awaking from a coma (r = 0.8823, P = 0.0100 in NMDAR group; r = 0.7906, P = 0.2000 in C-NORSE group). Pathways involved in humoral immune response, wound healing, and epigenetic regulation of transcription were upregulated in anti-NMDAR encephalitis NORSE. Pathways of innate and lymphocyte mediated immune response, synaptic functions, ubiquitination, and cell apoptosis were up-regulated in C-NORSE, which was consistent with a mouse model of status epilepticus. Fc receptor and B cell mediated immunity signaling pathways were downregulated in C-NORSE. Immunome microarray analysis demonstrated high autoantibody targeting 48 proteins in CSF samples of anti-NMDAR encephalitis NORSE. While the reaction was kept at a very low level in C-NORSE. There is no significant difference in inflammatory cytokine level between each group. The level of IL-4 (r = 0.7435, P = 0.0451), IL-13 (r = 0.7643, P = 0.0384), IFN-γ (r = 0.7973, P = 0.0287) and TNF-α (r = 0.8598, P = 0.0141) in NMDAR group, and IL-6 (r = 0.8479, P = 0.0348), IL-8 (r = 0.9076, P = 0.0166) in C-NORSE group were positively correlated with the up-proteomic score. The present study suggests that the up-proteomic score of CSF could be a promising indicator for assessment of the severity of anti-NMDAR encephalitis NORSE and C-NORSE. The distinct CSF proteomes imply different pathogenic mechanisms of the two diseases, and immunotherapy strategies as well.
Collapse
|
8
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
9
|
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
|
10
|
PAK1 Regulates MEC-17 Acetyltransferase Activity and Microtubule Acetylation during Proplatelet Extension. Int J Mol Sci 2020; 21:ijms21207531. [PMID: 33066011 PMCID: PMC7589885 DOI: 10.3390/ijms21207531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Mature megakaryocytes extend long processes called proplatelets from which platelets are released in the blood stream. The Rho GTPases Cdc42 and Rac as well as their downstream target, p21-activated kinase 2 (PAK2), have been demonstrated to be important for platelet formation. Here we address the role, during platelet formation, of PAK1, another target of the Rho GTPases. PAK1 decorates the bundled microtubules (MTs) of megakaryocyte proplatelets. Using a validated cell model which recapitulates proplatelet formation, elongation and platelet release, we show that lack of PAK1 activity increases the number of proplatelets but restrains their elongation. Moreover, in the absence of PAK1 activity, cells have hyperacetylated MTs and lose their MT network integrity. Using inhibitors of the tubulin deacetylase HDAC6, we demonstrate that abnormally high levels of MT acetylation are not sufficient to increase the number of proplatelets but cause loss of MT integrity. Taken together with our previous demonstration that MT acetylation is required for proplatelet formation, our data reveal that MT acetylation levels need to be tightly regulated during proplatelet formation. We identify PAK1 as a direct regulator of the MT acetylation levels during this process as we found that PAK1 phosphorylates the MT acetyltransferase MEC-17 and inhibits its activity.
Collapse
|
11
|
Ding XW, Li R, Geetha T, Tao YX, Babu JR. Nerve growth factor in metabolic complications and Alzheimer's disease: Physiology and therapeutic potential. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165858. [PMID: 32531260 DOI: 10.1016/j.bbadis.2020.165858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/11/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
As the population ages, obesity and metabolic complications as well as neurological disorders are becoming more prevalent, with huge economic burdens on both societies and families. New therapeutics are urgently needed. Nerve growth factor (NGF), first discovered in 1950s, is a neurotrophic factor involved in regulating cell proliferation, growth, survival, and apoptosis in both central and peripheral nervous systems. NGF and its precursor, proNGF, bind to TrkA and p75 receptors and initiate protein phosphorylation cascades, resulting in changes of cellular functions, and are associated with obesity, diabetes and its complications, and Alzheimer's disease. In this article, we summarize changes in NGF levels in metabolic and neuronal disorders, the signal transduction initiated by NGF and proNGF, the physiological and pathophysiological relevance, and therapeutic potential in treating chronic metabolic diseases and cognitive decline.
Collapse
Affiliation(s)
- Xiao-Wen Ding
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA
| | - Rongzi Li
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA
| | - Thangiah Geetha
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.
| | - Jeganathan Ramesh Babu
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
12
|
Patwa S, Benson CA, Dyer L, Olson K, Bangalore L, Hill M, Waxman SG, Tan AM. Spinal cord motor neuron plasticity accompanies second-degree burn injury and chronic pain. Physiol Rep 2019; 7:e14288. [PMID: 31858746 PMCID: PMC6923170 DOI: 10.14814/phy2.14288] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Burn injuries and associated complications present a major public health challenge. Many burn patients develop clinically intractable complications, including pain and other sensory disorders. Recent evidence has shown that dendritic spine neuropathology in spinal cord sensory and motor neurons accompanies central nervous system (CNS) or peripheral nervous system (PNS) trauma and disease. However, no research has investigated similar dendritic spine neuropathologies following a cutaneous thermal burn injury. In this retrospective investigation, we analyzed dendritic spine morphology and localization in alpha-motor neurons innervating a burn-injured area of the body (hind paw). To identify a molecular regulator of these dendritic spine changes, we further profiled motor neuron dendritic spines in adult mice treated with romidepsin, a clinically approved Pak1-inhibitor, or vehicle control at two postburn time points: Day 6 immediately after treatment, or Day 10 following drug withdrawal. In control treated mice, we observed an overall increase in dendritic spine density, including structurally mature spines with mushroom-shaped morphology. Pak1-inhibitor treatment reduced injury-induced changes to similar levels observed in animals without burn injury. The effectiveness of the Pak1-inhibitor was durable, since normalized dendritic spine profiles remained as long as 4 days despite drug withdrawal. This study is the first report of evidence demonstrating that a second-degree burn injury significantly affects motor neuron structure within the spinal cord. Furthermore, our results support the opportunity to study dendritic spine dysgenesis as a novel avenue to clarify the complexities of neurological disease following traumatic injury.
Collapse
Affiliation(s)
- Siraj Patwa
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Curtis A. Benson
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Lauren Dyer
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Kai‐Lan Olson
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Lakshmi Bangalore
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Myriam Hill
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Stephen G. Waxman
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| | - Andrew M. Tan
- Department of Neurology and Center for Neuroscience and Regeneration ResearchYale University School of MedicineNew HavenConnecticut
- Rehabilitation Research CenterVeterans Affairs Connecticut Healthcare SystemWest HavenConnecticut
| |
Collapse
|
13
|
Wu D, Wang S, Wen X, Han X, Wang Y, Shen M, Fan S, Zhuang J, Zhang Z, Shan Q, Li M, Hu B, Sun C, Lu J, Chen G, Zheng Y. Retracted
: Suppression of microRNA‐342‐3p increases glutamate transporters and prevents dopaminergic neuron loss through activating the Wnt signaling pathway via p21‐activated kinase 1 in mice with Parkinson's disease. J Cell Physiol 2018; 234:9033-9044. [DOI: 10.1002/jcp.27577] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 09/17/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Dong‐Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Xin‐Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Yong‐Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Shao‐Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- School of Environment Science and Spatial Informatics, China University of Mining and Technology Xuzhou China
- Jiangsu Key Laboratory for Eco‐Agricultural Biotechnology around Hongze Lake, School of Life Sciences, Huaiyin Normal University Huaian China
| | - Zi‐Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Meng‐Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Chun‐Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| | - Gui‐Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University Nanjing China
| | - Yuan‐Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University Xuzhou China
- College of Health Sciences, Jiangsu Normal University Xuzhou China
| |
Collapse
|
14
|
Sessa A, Ciabatti E, Drechsel D, Massimino L, Colasante G, Giannelli S, Satoh T, Akira S, Guillemot F, Broccoli V. The Tbr2 Molecular Network Controls Cortical Neuronal Differentiation Through Complementary Genetic and Epigenetic Pathways. Cereb Cortex 2018; 27:3378-3396. [PMID: 27600842 DOI: 10.1093/cercor/bhw270] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 08/04/2016] [Indexed: 01/21/2023] Open
Abstract
The T-box containing Tbr2 gene encodes for a transcription factor essential for the specification of the intermediate neural progenitors (INPs) originating the excitatory neurons of the cerebral cortex. However, its overall mechanism of action, direct target genes and cofactors remain unknown. Herein, we carried out global gene expression profiling combined with genome-wide binding site identification to determine the molecular pathways regulated by TBR2 in INPs. This analysis led to the identification of novel protein-protein interactions that control multiple features of INPs including cell-type identity, morphology, proliferation and migration dynamics. In particular, NEUROG2 and JMJD3 were found to associate with TBR2 revealing unexplored TBR2-dependent mechanisms. These interactions can explain, at least in part, the role of this transcription factor in the implementation of the molecular program controlling developmental milestones during corticogenesis. These data identify TBR2 as a major determinant of the INP-specific traits by regulating both genetic and epigenetic pathways.
Collapse
Affiliation(s)
- Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute , 20132 Milan, Italy
| | - Ernesto Ciabatti
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute , 20132 Milan, Italy
| | - Daniela Drechsel
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway ,LondonNW7 1AA, UK
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute , 20132 Milan, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute , 20132 Milan, Italy
| | - Serena Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute , 20132 Milan, Italy
| | - Takashi Satoh
- Laboratory of Host Defense, Osaka University, Osaka565-0871, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, Osaka University, Osaka565-0871, Japan
| | - Francois Guillemot
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway ,LondonNW7 1AA, UK
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.,CNR Institute of Neuroscience, 20129 Milan, Italy
| |
Collapse
|
15
|
Guo Y, Benson C, Hill M, Henry S, Effraim P, Waxman SG, Dib-Hajj S, Tan AM. Therapeutic potential of Pak1 inhibition for pain associated with cutaneous burn injury. Mol Pain 2018; 14:1744806918788648. [PMID: 29956587 PMCID: PMC6053256 DOI: 10.1177/1744806918788648] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 01/20/2023] Open
Abstract
Painful burn injuries are among the most debilitating form of trauma, globally ranking in the top 15 leading causes of chronic disease burden. Despite its prevalence, however, chronic pain after burn injury is under-studied. We previously demonstrated the contribution of the Rac1-signaling pathway in several models of neuropathic pain, including burn injury. However, Rac1 belongs to a class of GTPases with low therapeutic utility due to their complex intracellular dynamics. To further understand the mechanistic underpinnings of burn-induced neuropathic pain, we performed a longitudinal study to address the hypothesis that inhibition of the downstream effector of Rac1, Pak1, will improve pain outcome following a second-degree burn injury. Substantial evidence has identified Pak1 as promising a clinical target in cognitive dysfunction and is required for dendritic spine dysgenesis associated with many neurological diseases. In our burn injury model, mice exhibited significant tactile allodynia and heat hyperalgesia and dendritic spine dysgenesis in the dorsal horn. Activity-dependent expression of c-fos also increased in dorsal horn neurons, an indicator of elevated central nociceptive activity. To inhibit Pak1, we repurposed an FDA-approved inhibitor, romidepsin. Treatment with romidepsin decreased dendritic spine dysgenesis, reduced c-fos expression, and rescued pain thresholds. Drug discontinuation resulted in a relapse of cellular correlates of pain and in lower pain thresholds in behavioral tests. Taken together, our findings identify Pak1 signaling as a potential molecular target for therapeutic intervention in traumatic burn-induced neuropathic pain.
Collapse
Affiliation(s)
- Yiqun Guo
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Curtis Benson
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Myriam Hill
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Stefanie Henry
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Philip Effraim
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Sulayman Dib-Hajj
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| | - Andrew M Tan
- Department of Neurology, Center for Neuroscience and
Regeneration Research,
Yale
University School of Medicine, New
Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut
Healthcare System, West Haven, CT, USA
| |
Collapse
|
16
|
Inhibition of p21-Activated Kinase 1 by IPA-3 Promotes Locomotor Recovery After Spinal Cord Injury in Mice. Spine (Phila Pa 1976) 2016; 41:919-925. [PMID: 26863260 DOI: 10.1097/brs.0000000000001491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Ninety-six male adult CD-1 mice were randomly divided into sham, spinal cord injury (SCI) + vehicle, and SCI + IPA-3 groups. Expression of matrix metalloproteinase (MMP)-2 and MMP-9, production of tumor necrosis factors (TNF)-α and interleukin (IL)-1β, tissue edema, blood-spinal cord barrier penetrability, neural cell apoptosis, and neurological function recovery were measured. OBJECTIVE The aim of the study was to evaluate the effect of specific inhibition of p21-activated kinase 1 (PAK1) by IPA-3 on SCI and the underlying mechanisms thereof. SUMMARY OF BACKGROUND DATA SCI is a devastating clinical condition that may result in long-lasting and deteriorating functional deficits. The major goal of SCI treatment is to limit the development of secondary injury. IPA-3, a PAK1 inhibitor, exhibited neuroprotection against secondary damage after traumatic brain injury and subarachnoid hemorrhage (SAH). METHODS MMP-2, MMP-9, and cleaved caspase-3 expression were assessed by Western blot. Inflammatory cytokines TNF-α and IL-1β were detected by enzyme-linked immunosorbent assay (ELISA). The blood-spinal cord barrier disruption was measured by water content and Evans blue extravasation of the spinal cord. Neuronal apoptosis was evaluated by Nissl staining and Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling (TUNEL) assay. The locomotor behavior of hind limb was evaluated by Basso Mouse Scale (BMS) at 1, 3, 7, 14, and 28 days post-injury. RESULTS Compared with SCI + vehicle mice, IPA-3 treatment showed decreased p-PAK1, MMP-2, MMP-9, cleaved caspase-3, TNF-α, and IL-1β expression. Moreover, inhibition of PAK1 by IPA-3 reduced spinal cord water content and Evans blue extravasation, increased neuronal survival, and reduced TUNEL-positive cells at 24 hours after SCI. Furthermore, IPA-3 improved spinal cord functional recovery 7 days after SCI. CONCLUSION Inhibition of PAK1 by IPA-3 promoted recovery of neurological function, possibly by downregulating the expression of MMP-2, MMP-9, TNF-α, and IL-1β. Our data suggest that PAK1 may be a potential therapeutic target in patients with SCI. LEVEL OF EVIDENCE 1.
Collapse
|
17
|
nArgBP2 regulates excitatory synapse formation by controlling dendritic spine morphology. Proc Natl Acad Sci U S A 2016; 113:6749-54. [PMID: 27226294 DOI: 10.1073/pnas.1600944113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Neural Abelson-related gene-binding protein 2 (nArgBP2) was originally identified as a protein that directly interacts with synapse-associated protein 90/postsynaptic density protein 95-associated protein 3 (SAPAP3), a postsynaptic scaffolding protein critical for the assembly of glutamatergic synapses. Although genetic deletion of nArgBP2 in mice leads to manic/bipolar-like behaviors resembling many aspects of symptoms in patients with bipolar disorder, the actual function of nArgBP2 at the synapse is completely unknown. Here, we found that the knockdown (KD) of nArgBP2 by specific small hairpin RNAs (shRNAs) resulted in a dramatic change in dendritic spine morphology. Reintroducing shRNA-resistant nArgBP2 reversed these defects. In particular, nArgBP2 KD impaired spine-synapse formation such that excitatory synapses terminated mostly at dendritic shafts instead of spine heads in spiny neurons, although inhibitory synapse formation was not affected. nArgBP2 KD further caused a marked increase of actin cytoskeleton dynamics in spines, which was associated with increased Wiskott-Aldrich syndrome protein-family verprolin homologous protein 1 (WAVE1)/p21-activated kinase (PAK) phosphorylation and reduced activity of cofilin. These effects of nArgBP2 KD in spines were rescued by inhibiting PAK or activating cofilin combined with sequestration of WAVE. Together, our results suggest that nArgBP2 functions to regulate spine morphogenesis and subsequent spine-synapse formation at glutamatergic synapses. They also raise the possibility that the aberrant regulation of synaptic actin filaments caused by reduced nArgBP2 expression may contribute to the manifestation of the synaptic dysfunction observed in manic/bipolar disorder.
Collapse
|
18
|
Singh NK, Kotla S, Dyukova E, Traylor JG, Orr AW, Chernoff J, Marion TN, Rao GN. Disruption of p21-activated kinase 1 gene diminishes atherosclerosis in apolipoprotein E-deficient mice. Nat Commun 2015; 6:7450. [PMID: 26104863 PMCID: PMC4480433 DOI: 10.1038/ncomms8450] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 05/09/2015] [Indexed: 12/26/2022] Open
Abstract
Pak1 plays an important role in various cellular processes, including cell motility, polarity, survival and proliferation. To date, its role in atherogenesis has not been explored. Here we report the effect of Pak1 on atherogenesis using atherosclerosis-prone apolipoprotein E-deficient (ApoE−/−) mice as a model. Disruption of Pak1 in ApoE−/− mice results in reduced plaque burden, significantly attenuates circulating IL-6 and MCP-1 levels, limits the expression of adhesion molecules and diminishes the macrophage content in the aortic root of ApoE−/− mice. We also observed reduced oxidized LDL uptake and increased cholesterol efflux by macrophages and smooth muscle cells of ApoE−/−:Pak1−/− mice as compared with ApoE−/− mice. In addition, we detect increased Pak1 phosphorylation in human atherosclerotic arteries, suggesting its role in human atherogenesis. Altogether, these results identify Pak1 as an important factor in the initiation and progression of atherogenesis. Atherogenesis involves coordinated action of different cell types and factors. Here the authors show that the kinase Pak1 represents a key pro-atherogenic factor affecting the function of macrophages and vascular smooth muscle cells, including their production of proinflammatory cytokine IL-6 and chemokine MCP-1, and retention of cholesterol.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| | - Sivareddy Kotla
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| | - Elena Dyukova
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| | - James G Traylor
- Department of Pathology, LSU Health Sciences Center, Shreveport, Louisiana 71103, USA
| | - A Wayne Orr
- Department of Pathology, LSU Health Sciences Center, Shreveport, Louisiana 71103, USA
| | - Jonathan Chernoff
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, Pennsylvania 19111, USA
| | - Tony N Marion
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis, Tennessee 38163, USA
| |
Collapse
|
19
|
Pan X, Chang X, Leung C, Zhou Z, Cao F, Xie W, Jia Z. PAK1 regulates cortical development via promoting neuronal migration and progenitor cell proliferation. Mol Brain 2015; 8:36. [PMID: 26043730 PMCID: PMC4456803 DOI: 10.1186/s13041-015-0124-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/12/2015] [Indexed: 11/17/2022] Open
Abstract
Background p21-activated kinase 1 (PAK1) is a serine/threonine kinase known to be activated by the Rho family small GTPases and to play a key role in cytoskeletal reorganization, spine morphology and synaptic plasticity. PAK1 is also implicated in a number of neurodevelopmental and neurodegenerative diseases, including autism, intellectual disability and Alzheimer’s disease. However, the role of PAK1 in early brain development remains unknown. Results In this study, we employed genetic manipulations to investigate the role of PAK1 in the cerebral cortical development in mice. We showed that compared to the wild type littermates, PAK1 knockout mice have a reduction in the number of pyramidal neurons in several layers of the cerebral cortex, which is associated with a smaller pool of neural progenitor cells and impaired neuronal migration. Conclusion These results suggest that PAK1 regulates cortical development by promoting the proliferation of neural progenitor cells and facilitating the migration of these neurons to specific regions of the cortex.
Collapse
Affiliation(s)
- Xingxiu Pan
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Xinxia Chang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Celeste Leung
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Zikai Zhou
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Feng Cao
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China. .,Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, 2 Sipailou Road, 210096, Nanjing, China.
| | - Zhengping Jia
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
20
|
Clinicopathological and cellular signature of PAK1 in human bladder cancer. Tumour Biol 2014; 36:2359-68. [PMID: 25412958 DOI: 10.1007/s13277-014-2843-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/12/2014] [Indexed: 02/08/2023] Open
Abstract
Bladder cancer (BC) is the ninth most common cancer and the 13th most common cause of cancer death. Although p21 protein-activated kinase (PAK) regulates cell growth, motility, and morphology, the expression and function of PAK1 associated with the clinicopathological and cellular signature of human BC are not clear. This study was to examine the expression of PAK1 in human BC, the association of PAK1 with clinicopathological features, and the effect of PAK1 on cell proliferation, migration, and invasion in BC cells. A total of 54 BC and 12 normal bladder tissue specimens were retrieved. Among 54 BC patients, 39 cases were superficial BC and 15 cases were invasive BC. Histological examination revealed 29 patients with low-grade and 25 patients with high-grade papillary urothelial carcinomas. Immunohistochemical staining showed that PAK1 was overexpressed in BC tissue compared with normal bladder tissue. The overexpression of PAK1 was significantly associated with tumor size, histological grade, and lymph node metastasis, but not with gender, age, clinical stage, tumor number, and recurrence. Furthermore, the cytoplasmic distribution of PAK1 was observed in BC cells. Knocking down of PAK1 using lentiviral transduction decreased BC cell proliferation, migration, and invasion. In conclusion, we demonstrated that the overexpression of PAK1 is closely associated with the clinicopathological features of BC, suggesting that PAK1 may play an important role in the development and progression of BC and may be a potential therapeutic target for the treatment of BC.
Collapse
|
21
|
Inhibition of p21-activated kinase 1 by IPA-3 attenuates secondary injury after traumatic brain injury in mice. Brain Res 2014; 1585:13-22. [PMID: 25148711 DOI: 10.1016/j.brainres.2014.08.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/26/2023]
Abstract
The p21-activated kinase 1 (PAK1) is up-regulated in the brain following traumatic brain injury (TBI). Inhibition of PAK1 has been found to alleviate brain edema in a rat model of subarachnoid hemorrhage. Suppressing PAK1 activity might represent a novel therapeutics of attenuating secondary injury following TBI. Here we confirmed that the mRNA and protein levels of PAK1 and the protein level of p-PAK1 were significantly increased after inducing TBI in mice via M.A. Flierl's weight-drop model. A single intraperitoneal administration of IPA-3, a specific PAK1 inhibitor, immediately after TBI significantly reduced the protein level of p-PAK1, cleaved caspase-3 level, the number of apoptotic cells at the lesion sites of TBI mice. It also reduced brain water content and the blood-brain barrier permeability in TBI mice. Furthermore, the administration of IPA-3 significantly reduced the neurological severity score and increased the grip test score in TBI mice. Taken together, we demonstrate that PAK1 inhibition by IPA-3 may attenuate the secondary injury following TBI, suggesting it might be a promising neuroprotective strategy for preventing the development of secondary injury after TBI.
Collapse
|
22
|
Um K, Niu S, Duman JG, Cheng JX, Tu YK, Schwechter B, Liu F, Hiles L, Narayanan AS, Ash RT, Mulherkar S, Alpadi K, Smirnakis SM, Tolias KF. Dynamic control of excitatory synapse development by a Rac1 GEF/GAP regulatory complex. Dev Cell 2014; 29:701-15. [PMID: 24960694 DOI: 10.1016/j.devcel.2014.05.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 04/23/2014] [Accepted: 05/14/2014] [Indexed: 11/18/2022]
Abstract
The small GTPase Rac1 orchestrates actin-dependent remodeling essential for numerous cellular processes including synapse development. While precise spatiotemporal regulation of Rac1 is necessary for its function, little is known about the mechanisms that enable Rac1 activators (GEFs) and inhibitors (GAPs) to act in concert to regulate Rac1 signaling. Here, we identify a regulatory complex composed of a Rac-GEF (Tiam1) and a Rac-GAP (Bcr) that cooperate to control excitatory synapse development. Disruption of Bcr function within this complex increases Rac1 activity and dendritic spine remodeling, resulting in excessive synaptic growth that is rescued by Tiam1 inhibition. Notably, EphB receptors utilize the Tiam1-Bcr complex to control synaptogenesis. Following EphB activation, Tiam1 induces Rac1-dependent spine formation, whereas Bcr prevents Rac1-mediated receptor internalization, promoting spine growth over retraction. The finding that a Rac-specific GEF/GAP complex is required to maintain optimal levels of Rac1 signaling provides an important insight into the regulation of small GTPases.
Collapse
Affiliation(s)
- Kyongmi Um
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sanyong Niu
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Joseph G Duman
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jinxuan X Cheng
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yen-Kuei Tu
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Brandon Schwechter
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Feng Liu
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Laura Hiles
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Anjana S Narayanan
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ryan T Ash
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Kannan Alpadi
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Stelios M Smirnakis
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
23
|
Abstract
p21-Activated protein kinases (PAKs) are centrally involved in a plethora of cellular processes and functions. Their function as effectors of small GTPases Rac1 and Cdc42 has been extensively studied during the past two decades, particularly in the realms of cell proliferation, apoptosis, and hence tumorigenesis, as well as cytoskeletal remodeling and related cellular events in health and disease. In recent years, a large number of studies have shed light onto the fundamental role of group I PAKs, most notably PAK1, in metabolic homeostasis. In skeletal muscle, PAK1 was shown to mediate the function of insulin on stimulating GLUT4 translocation and glucose uptake, while in pancreatic β-cells, PAK1 participates in insulin granule localization and vesicle release. Furthermore, we demonstrated that PAK1 mediates the cross talk between insulin and Wnt/β-catenin signaling pathways and hence regulates gut proglucagon gene expression and the production of the incretin hormone glucagon-like peptide-1 (GLP-1). The utilization of chemical inhibitors of PAK and the characterization of Pak1(-/-) mice enabled us to gain mechanistic insights as well as to assess the overall contribution of PAKs in metabolic homeostasis. This review summarizes our current understanding of PAKs, with an emphasis on the emerging roles of PAK1 in glucose homeostasis.
Collapse
|
24
|
Zhang D, Pekkanen-Mattila M, Shahsavani M, Falk A, Teixeira AI, Herland A. A 3D Alzheimer's disease culture model and the induction of P21-activated kinase mediated sensing in iPSC derived neurons. Biomaterials 2014; 35:1420-8. [DOI: 10.1016/j.biomaterials.2013.11.028] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/08/2013] [Indexed: 12/22/2022]
|
25
|
p21-Activated kinase (PAK) is required for Bone Morphogenetic Protein (BMP)-induced dendritogenesis in cortical neurons. Mol Cell Neurosci 2013; 57:83-92. [PMID: 24141051 DOI: 10.1016/j.mcn.2013.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 10/01/2013] [Accepted: 10/08/2013] [Indexed: 01/21/2023] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are crucial for many aspects of the development and differentiation of the nervous system and are important in controlling cytoskeletal remodeling during neuronal morphogenesis. BMPs are TGFβ superfamily members that signal through a heteromeric complex of type I and type II BMP receptors. The BMPRII receptor is particularly important in mediating remodeling of the neuronal cytoskeleton through the activation of BMPRII-bound cytoskeletal regulators, such as LIM Kinase (LIMK). Here, we show that PAK1, a key regulator of diverse neuronal processes and an upstream activator of LIMK, binds to the BMP type I receptor, ALK2. Although, PAK1 is dispensable for activation of the Smad transcriptional mediators, abrogation of PAK1 expression or inhibition of PAK1 activity prevents BMP-induced neurite outgrowth in cultured neuroblastoma cell lines. Moreover, in primary murine embryonic cortical neurons, inhibition of PAK activity blocks BMP7-induced cofilin phosphorylation, prevents remodeling of the actin cytoskeleton and thereby blocks BMP7-induced dendrite formation. Thus, we propose a model in which BMP7 signaling leads to the recruitment of ALK2-bound PAK1 to BMPRII, which binds a downstream regulator of the actin cytoskeleton, LIMK1, and that the BMP receptor complex thereby acts as a scaffold to localize and coordinate actin cytoskeletal remodeling. We propose that this scaffold plays a key role in mediating BMP7-dependent dendritogenesis in primary cortical neurons.
Collapse
|
26
|
Chandrasekaran S, Bonchev D. A network view on Parkinson's disease. Comput Struct Biotechnol J 2013; 7:e201304004. [PMID: 24688734 PMCID: PMC3962195 DOI: 10.5936/csbj.201304004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/27/2013] [Accepted: 06/30/2013] [Indexed: 12/21/2022] Open
Abstract
Network-based systems biology tools including Pathway Studio 9.0 were used to identify Parkinson's disease (PD) critical molecular players, drug targets, and underlying biological processes. Utilizing several microarray gene expression datasets, biomolecular networks such as direct interaction, shortest path, and microRNA regulatory networks were constructed and analyzed for the disease conditions. Network topology analysis of node connectivity and centrality revealed in combination with the guilt-by-association rule 17 novel genes of PD-potential interest. Seven new microRNAs (miR-132, miR-133a1, miR-181-1, miR-182, miR-218-1, miR-29a, and miR-330) related to Parkinson's disease were identified, along with more microRNA targeted genes of interest like RIMS3, SEMA6D and SYNJ1. David and IPA enrichment analysis of KEGG and canonical pathways provided valuable mechanistic information emphasizing among others the role of chemokine signaling, adherence junction, and regulation of actin cytoskeleton pathways. Several routes for possible disease initiation and neuro protection mechanisms triggered via the extra-cellular ligands such as CX3CL1, SEMA6D and IL12B were thus uncovered, and a dual regulatory system of integrated transcription factors and microRNAs mechanisms was detected.
Collapse
Affiliation(s)
- Sreedevi Chandrasekaran
- Center for the Study of Biological Complexity, Virginia Commonwealth University, United States
| | - Danail Bonchev
- Center for the Study of Biological Complexity, Virginia Commonwealth University, United States
| |
Collapse
|
27
|
Arsenault D, Dal-Pan A, Tremblay C, Bennett DA, Guitton MJ, De Koninck Y, Tonegawa S, Calon F. PAK inactivation impairs social recognition in 3xTg-AD Mice without increasing brain deposition of tau and Aβ. J Neurosci 2013; 33:10729-40. [PMID: 23804095 PMCID: PMC4019789 DOI: 10.1523/jneurosci.1501-13.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/13/2013] [Indexed: 11/21/2022] Open
Abstract
Defects in p21-activated kinase (PAK) are suspected to play a role in cognitive symptoms of Alzheimer's disease (AD). Dysfunction in PAK leads to cofilin activation, drebrin displacement from its actin-binding site, actin depolymerization/severing, and, ultimately, defects in spine dynamics and cognitive impairment in mice. To determine the role of PAK in AD, we first quantified PAK by immunoblotting in homogenates from the parietal neocortex of subjects with a clinical diagnosis of no cognitive impairment (n = 12), mild cognitive impairment (n = 12), or AD (n = 12). A loss of total PAK, detected in the cortex of AD patients (-39% versus controls), was correlated with cognitive impairment (r(2) = 0.148, p = 0.027) and deposition of total and phosphorylated tau (r(2) = 0.235 and r(2) = 0.206, respectively), but not with Aβ42 (r(2) = 0.056). Accordingly, we found a decrease of total PAK in the cortex of 12- and 20-month-old 3xTg-AD mice, an animal model of AD-like Aβ and tau neuropathologies. To determine whether PAK dysfunction aggravates AD phenotype, 3xTg-AD mice were crossed with dominant-negative PAK mice. PAK inactivation led to obliteration of social recognition in old 3xTg-AD mice, which was associated with a decrease in cortical drebrin (-25%), but without enhancement of Aβ/tau pathology or any clear electrophysiological signature. Overall, our data suggest that PAK decrease is a consequence of AD neuropathology and that therapeutic activation of PAK may exert symptomatic benefits on high brain function.
Collapse
Affiliation(s)
- Dany Arsenault
- Faculté de pharmacie, Université Laval, Quebec City, G1V 0A6, Quebec, Canada
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| | - Alexandre Dal-Pan
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| | - Cyntia Tremblay
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
| | - Matthieu J. Guitton
- Faculté de médecine, Université Laval, Quebec City, Quebec, G1V 0A6, Canada
- Institut Universitaire en Santé Mentale de Québec, Quebec City, Quebec, G1J 2G3, Canada, and
| | - Yves De Koninck
- Faculté de médecine, Université Laval, Quebec City, Quebec, G1V 0A6, Canada
- Institut Universitaire en Santé Mentale de Québec, Quebec City, Quebec, G1J 2G3, Canada, and
| | - Susumu Tonegawa
- Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec City, G1V 0A6, Quebec, Canada
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| |
Collapse
|
28
|
Wada-Kiyama Y, Suzuki C, Hamada T, Rai D, Kiyama R, Kaneda M, Sakuma Y. Estrogen-induced cell signaling in the sexually dimorphic nucleus of the rat preoptic area: potential involvement of cofilin in actin dynamics for cell migration. Biochem Biophys Res Commun 2013; 434:287-92. [PMID: 23537649 DOI: 10.1016/j.bbrc.2013.02.117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 02/24/2013] [Indexed: 10/27/2022]
Abstract
Estrogen is a key factor to induce the sexually dimorphic nucleus (SDN) in the preoptic area (POA) of the rat brain. Identification of estrogen-dependent signaling pathways at SDN in POA during the critical period is a prerequisite for elucidating the mechanism. In the present study, we treated female rats with/without 17β-estradiol (E2) at birth, designated as postnatal day 1 (P1), and prepared total RNA from brain slices containing SDN for DNA microarray analysis. Among the estrogen-responsive genes identified, protein kinase C-delta (PKC-δ) was significantly up-regulated by E2 at P5. We examined the downstream effectors of PKC-δ protein by Western blotting and found an E2-induced PKC-δ/Rac1/PAK1/LIMK1/cofilin pathway. In the pathway, E2 suppressed the phosphorylation (inactive form) of cofilin. This result was supported by immunohistochemistry, where the phosphorylation/dephosphorylation of cofilin occurred at SDN, which suggests that cell migration is a cue to create sexual dimorphism in POA.
Collapse
Affiliation(s)
- Yuko Wada-Kiyama
- Department of Physiology, Nippon Medical School, Tokyo 113-8602, Japan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Asrar S, Jia Z. Molecular mechanisms coordinating functional and morphological plasticity at the synapse: Role of GluA2/N-cadherin interaction-mediated actin signaling in mGluR-dependent LTD. Cell Signal 2013; 25:397-402. [DOI: 10.1016/j.cellsig.2012.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 11/05/2012] [Indexed: 11/25/2022]
|
30
|
Penzes P, Cahill ME. Deconstructing signal transduction pathways that regulate the actin cytoskeleton in dendritic spines. Cytoskeleton (Hoboken) 2012; 69:426-41. [PMID: 22307832 DOI: 10.1002/cm.21015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 11/10/2022]
Abstract
Dendritic spines are the sites of most excitatory synapses in the central nervous system. Recent studies have shown that spines function independently of each other, and they are currently the smallest known processing units in the brain. Spines exist in an array of morphologies, and spine structure helps dictate synaptic function. Dendritic spines are rich in actin, and actin rearrangements are critical regulators of spine morphology and density. In this review, we discuss the importance of actin in regulating dendritic spine morphogenesis, and discuss the upstream signal transduction pathways that either foster or inhibit actin polymerization. The understanding of actin regulatory pathways is best conceptualized as a hierarchical network in which molecules function in discrete levels defined by their molecular distance to actin. To this end, we focus on several classes of molecules, including guanine nucleotide exchange factors, small GTPases, small GTPase effectors, and actin binding proteins. We discuss how individual proteins in these molecular classes impact spine morphogenesis, and reveal the biochemical interactions in these networks that are responsible for shaping actin polymerization. Finally, we discuss the importance of these actin regulatory pathways in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | | |
Collapse
|
31
|
The secret life of kinases: functions beyond catalysis. Cell Commun Signal 2011; 9:23. [PMID: 22035226 PMCID: PMC3215182 DOI: 10.1186/1478-811x-9-23] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 10/28/2011] [Indexed: 02/07/2023] Open
Abstract
Protein phosphorylation participates in the regulation of all fundamental biological processes, and protein kinases have been intensively studied. However, while the focus was on catalytic activities, accumulating evidence suggests that non-catalytic properties of protein kinases are essential, and in some cases even sufficient for their functions. These non-catalytic functions include the scaffolding of protein complexes, the competition for protein interactions, allosteric effects on other enzymes, subcellular targeting, and DNA binding. This rich repertoire often is used to coordinate phosphorylation events and enhance the specificity of substrate phosphorylation, but also can adopt functions that do not rely on kinase activity. Here, we discuss such kinase independent functions of protein and lipid kinases focussing on kinases that play a role in the regulation of cell proliferation, differentiation, apoptosis, and motility.
Collapse
|
32
|
Raman D, Milatovic SZ, Milatovic D, Splittgerber R, Fan GH, Richmond A. Chemokines, macrophage inflammatory protein-2 and stromal cell-derived factor-1α, suppress amyloid β-induced neurotoxicity. Toxicol Appl Pharmacol 2011; 256:300-13. [PMID: 21704645 DOI: 10.1016/j.taap.2011.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 06/08/2011] [Accepted: 06/09/2011] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is characterized by a progressive cognitive decline and accumulation of neurotoxic oligomeric peptides amyloid-β (Aβ). Although the molecular events are not entirely known, it has become evident that inflammation, environmental and other risk factors may play a causal, disruptive and/or protective role in the development of AD. The present study investigated the ability of the chemokines, macrophage inflammatory protein-2 (MIP-2) and stromal cell-derived factor-1α (SDF-1α), the respective ligands for chemokine receptors CXCR2 and CXCR4, to suppress Aβ-induced neurotoxicity in vitro and in vivo. Pretreatment with MIP-2 or SDF-1α significantly protected neurons from Aβ-induced dendritic regression and apoptosis in vitro through activation of Akt, ERK1/2 and maintenance of metalloproteinase ADAM17 especially with SDF-1α. Intra-cerebroventricular (ICV) injection of Aβ led to reduction in dendritic length and spine density of pyramidal neurons in the CA1 area of the hippocampus and increased oxidative damage 24h following the exposure. The Aβ-induced morphometric changes of neurons and increase in biomarkers of oxidative damage, F(2)-isoprostanes, were significantly inhibited by pretreatment with the chemokines MIP-2 or SDF-1α. Additionally, MIP-2 or SDF-1α was able to suppress the aberrant mislocalization of p21-activated kinase (PAK), one of the proteins involved in the maintenance of dendritic spines. Furthermore, MIP-2 also protected neurons against Aβ neurotoxicity in CXCR2-/- mice, potentially through observed up regulation of CXCR1 mRNA. Understanding the neuroprotective potential of chemokines is crucial in defining the role for their employment during the early stages of neurodegeneration.
Collapse
Affiliation(s)
- Dayanidhi Raman
- Department of Cancer Biology, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
33
|
Murakoshi H, Wang H, Yasuda R. Local, persistent activation of Rho GTPases during plasticity of single dendritic spines. Nature 2011; 472:100-4. [PMID: 21423166 PMCID: PMC3105377 DOI: 10.1038/nature09823] [Citation(s) in RCA: 415] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Accepted: 01/13/2011] [Indexed: 12/15/2022]
Abstract
The Rho family of GTPases play important roles in morphogenesis of dendritic spines1–3 and synaptic plasticity4–9 by modulating the organization of the actin cytoskeleton10. Here, we monitored the activity of Rho GTPases, RhoA and Cdc42, in single dendritic spines undergoing structural plasticity associated with long-term potentiation (LTP) using 2-photon fluorescence lifetime imaging microscopy (2pFLIM)11–13. When long-term volume increase was induced in a single spine using 2-photon glutamate uncaging14,15, RhoA and Cdc42 were rapidly activated in the stimulated spine. These activities decayed over ~5 min, and were then followed by a phase of persistent activation lasting more than 30 min. Although active RhoA and Cdc42 were similarly mobile, their activity patterns were different. RhoA activation diffused out of the stimulated spine and spread over ~5 μm along the dendrite. In contrast, Cdc42 activation was restricted to the stimulated spine, and exhibited a steep gradient at the spine necks. Inhibition of the Rho-Rock pathway preferentially inhibited the initial spine growth, whereas the inhibition of the Cdc42-Pak pathway blocked the maintenance of sustained structural plasticity. RhoA and Cdc42 activation depended on Ca2+/calmodulin-dependent kinase (CaMKII). Thus, RhoA and Cdc42 relay transient CaMKII activation13 to synapse-specific, long-term signalling required for spine structural plasticity.
Collapse
Affiliation(s)
- Hideji Murakoshi
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
34
|
p21-Activated kinases 1 and 3 control brain size through coordinating neuronal complexity and synaptic properties. Mol Cell Biol 2010; 31:388-403. [PMID: 21115725 DOI: 10.1128/mcb.00969-10] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The molecular mechanisms that coordinate postnatal brain enlargement, synaptic properties, and cognition remain an enigma. Here, we demonstrate that neuronal complexity controlled by p21-activated kinases (PAKs) is a key determinant for postnatal brain enlargement and synaptic properties. We showed that double-knockout (DK) mice lacking both PAK1 and PAK3 were born healthy, with normal brain size and structure, but severely impaired in postnatal brain growth, resulting in a dramatic reduction in brain volume. Remarkably, the reduced brain size was accompanied by minimal changes in total cell count, due to a significant increase in cell density. However, the DK neurons have smaller soma, markedly simplified dendritic arbors/axons, and reduced synapse density. Surprisingly, the DK mice had elevated basal synaptic responses due to enhanced individual synaptic potency but were severely impaired in bidirectional synaptic plasticity. The actions of PAK1 and PAK3 are possibly mediated by cofilin-dependent actin regulation, because the activity of cofilin and the properties of actin filaments were altered in the DK mice. These results reveal an essential in vivo role of PAK1 and PAK3 in coordinating neuronal complexity and synaptic properties and highlight the critical importance of dendrite/axon growth in dictating postnatal brain growth and attainment of normal brain size and function.
Collapse
|
35
|
de la Torre-Ubieta L, Gaudillière B, Yang Y, Ikeuchi Y, Yamada T, DiBacco S, Stegmüller J, Schüller U, Salih DA, Rowitch D, Brunet A, Bonni A. A FOXO-Pak1 transcriptional pathway controls neuronal polarity. Genes Dev 2010; 24:799-813. [PMID: 20395366 DOI: 10.1101/gad.1880510] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neuronal polarity is essential for normal brain development and function. However, cell-intrinsic mechanisms that govern the establishment of neuronal polarity remain to be identified. Here, we report that knockdown of endogenous FOXO proteins in hippocampal and cerebellar granule neurons, including in the rat cerebellar cortex in vivo, reveals a requirement for the FOXO transcription factors in the establishment of neuronal polarity. The FOXO transcription factors, including the brain-enriched protein FOXO6, play a critical role in axo-dendritic polarization of undifferentiated neurites, and hence in a switch from unpolarized to polarized neuronal morphology. We also identify the gene encoding the protein kinase Pak1, which acts locally in neuronal processes to induce polarity, as a critical direct target gene of the FOXO transcription factors. Knockdown of endogenous Pak1 phenocopies the effect of FOXO knockdown on neuronal polarity. Importantly, exogenous expression of Pak1 in the background of FOXO knockdown in both primary neurons and postnatal rat pups in vivo restores the polarized morphology of neurons. These findings define the FOXO proteins and Pak1 as components of a cell-intrinsic transcriptional pathway that orchestrates neuronal polarity, thus identifying a novel function for the FOXO transcription factors in a unique aspect of neural development.
Collapse
|
36
|
Dorsam ST, Vomhof-Dekrey E, Hermann RJ, Haring JS, Van der Steen T, Wilkerson E, Boskovic G, Denvir J, Dementieva Y, Primerano D, Dorsam GP. Identification of the early VIP-regulated transcriptome and its associated, interactome in resting and activated murine CD4 T cells. Mol Immunol 2010; 47:1181-94. [PMID: 20117839 DOI: 10.1016/j.molimm.2010.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 12/15/2009] [Accepted: 01/05/2010] [Indexed: 01/12/2023]
Abstract
More than 40 years after the discovery of vasoactive intestinal peptide (VIP), its transcriptome in the immune system has still not been completely elucidated. In an attempt to understand the biological role of this neuropeptide in immunity, we chose CD4 T cells as a cellular system. Agilent Mouse Whole Genome microarrays were hybridized with fluorescently labeled total RNA isolated from resting CD4 T cells cultured +/-10(-7)M VIP for 5h or PMA/ionomycin activated CD4 T cells cultured +/-10(-7)M VIP for 5h. These VIP-regulated transcriptomes were analyzed by Significance Analysis of Microarrays (SAM) and Ingenuity Pathway Analysis (IPA) software to identify relevant signaling pathways modulated by VIP in the absence and presence of T cell activation. In resting CD4 T cells, VIP-modulated 368 genes, ranging from 3.49 to -4.78-fold. In the PMA/ionomycin activated CD4 T cells, 326 gene expression levels were changed by VIP, ranging from 2.94 to -1.66-fold. IPA analysis revealed that VIP exposure alters cellular function through EGFR signaling in resting CD4 T cells, and modulates immediate early genes, Fos and CREM/ICER, in activated CD4 T cells. These gene expression changes are suggested to explain at a molecular level how VIP can regulate T cell homing to the gut and induce regulatory T cell generation.
Collapse
Affiliation(s)
- Sheri Tinnell Dorsam
- Department of Chemistry and Molecular Biology, North Dakota State University, Fargo, ND 58108-6050, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chico LK, Van Eldik LJ, Watterson DM. Targeting protein kinases in central nervous system disorders. Nat Rev Drug Discov 2009; 8:892-909. [PMID: 19876042 PMCID: PMC2825114 DOI: 10.1038/nrd2999] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein kinases are a growing drug target class in disorders in peripheral tissues, but the development of kinase-targeted therapies for central nervous system (CNS) diseases remains a challenge, largely owing to issues associated specifically with CNS drug discovery. However, several candidate therapeutics that target CNS protein kinases are now in various stages of preclinical and clinical development. We review candidate compounds and discuss selected CNS protein kinases that are emerging as important therapeutic targets. In addition, we analyse trends in small-molecule properties that correlate with key challenges in CNS drug discovery, such as blood-brain barrier penetrance and cytochrome P450-mediated metabolism, and discuss the potential of future approaches that will integrate molecular-fragment expansion with pharmacoinformatics to address these challenges.
Collapse
Affiliation(s)
- Laura K Chico
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
38
|
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: 28] [Impact Index Per Article: 1.8] [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
|
39
|
A novel function for p53: regulation of growth cone motility through interaction with Rho kinase. J Neurosci 2009; 29:5183-92. [PMID: 19386914 DOI: 10.1523/jneurosci.0420-09.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The transcription factor p53 suppresses tumorgenesis by regulating cell proliferation and migration. We investigated whether p53 could also control cell motility in postmitotic neurons. p53 isoforms recognized by phospho-p53-specific (at Ser-15) or "mutant" conformation-specific antibodies were highly and specifically expressed in axons and axonal growth cones in primary hippocampal neurons. Inhibition of p53 function by inhibitors, small interfering RNAs, or by dominant-negative forms, induced axonal growth cone collapse, whereas p53 overexpression led to larger growth cones. Furthermore, deletion of the p53 nuclear export signal blocked its axonal distribution and induced growth cone collapse. p53 inhibition-induced axonal growth cone collapse was significantly reduced by the Rho kinase (ROCK) inhibitor, Y27632 [(R)-(+)-trans-N-(4-pyridyl)-4-(1-aminoethyl)-cyclohexanecarboxamide]. Our results reveal a new function for p53 as a critical regulator of axonal growth cone behavior by suppressing ROCK activity.
Collapse
|
40
|
p21-Activated kinase mediates rapid estradiol-negative feedback actions in the reproductive axis. Proc Natl Acad Sci U S A 2009; 106:7221-6. [PMID: 19359483 DOI: 10.1073/pnas.0812597106] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Nonclassical estrogen receptor alpha (ERalpha) signaling can mediate E(2) negative feedback actions in the reproductive axis; however, downstream pathways conveying these effects remain unclear. These studies tested the hypothesis that p21-activated kinase 1 (PAK1), a serine/threonine kinase rapidly activated by E(2) in nonneural cells, functions as a downstream node for E(2) signaling pathways in cells of the preoptic area, and it may thereby mediate E(2) negative feedback effects. Treatment of ovariectomized (OVX) rats with estradiol benzoate (EB) caused rapid and transient induction of phosphorylated PAK1 immunoreactivity in the medial preoptic nucleus (MPN) but not the arcuate nucleus. To determine whether rapid induction of PAK phosphorylation by E(2) is mediated by nonclassical [estrogen response element (ERE)-independent] ERalpha signaling, we used female ERalpha null (ERalpha(-/-)) mice possessing an ER knock-in mutation (E207A/G208A; AA), in which the mutant ERalpha is incapable of binding DNA and can signal only through membrane-initiated or ERE-independent genotropic pathways (ERalpha(-/AA) mice). After 1-h EB treatment, the number of pPAK1-immunoreactive cells in the MPN was increased in both wild-type (ERalpha(+/+)) and ERalpha(-/AA) mice but was unchanged in ERalpha(-/-) mice. Serum luteinizing hormone (LH) was likewise suppressed within 1 h after EB treatment in ERalpha(+/+) and ERalpha(-/AA) but not ERalpha(-/ -) mice. In OVX rats, 5-min intracerebroventricular infusion of a PAK inhibitor peptide but not control peptide blocked rapid EB suppression of LH secretion. Taken together, our findings implicate PAK1 activation subsequent to nonclassical ERalpha signaling as an important component of the negative feedback actions of E(2) in the brain.
Collapse
|