1
|
Gourisankar S, Nettles SA, Wenderski W, Paulo JA, Kim SH, Roepke KC, Ellis C, Abuzaid HZ, Gygi SP, Crabtree GR. Synaptic activity causes minute-scale changes in BAF complex composition and function. Mol Cell 2025:S1097-2765(25)00456-3. [PMID: 40494354 DOI: 10.1016/j.molcel.2025.05.017] [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: 10/24/2023] [Revised: 04/02/2025] [Accepted: 05/14/2025] [Indexed: 06/18/2025]
Abstract
Genes encoding subunits of the BAF ATP-dependent chromatin remodeling complex are among the most enriched for deleterious de novo mutations in intellectual disabilities and autism spectrum disorder, but the causative molecular pathways are not understood. Synaptic activity in neurons is critical for learning, memory, and proper neural development. While BAF is required for activity-dependent developmental processes, such as dendritic outgrowth, the immediate molecular consequences of neuronal activity on BAF complexes are unknown. Here, we report that neuronal activity induces dramatic remodeling of the subunit composition of BAF complexes within 15 min, concurrent with both phosphorylation and dephosphorylation of its subunits. These biochemical effects are a convergent phenomenon downstream of multiple calcium-activated signaling pathways in mouse neurons and mouse fibroblasts and correspond to changes in BAF-dependent chromatin accessibility. Our studies imply that BAF decodes signals at the membrane by altering the combinatorial composition of its subunits.
Collapse
Affiliation(s)
- Sai Gourisankar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Sabin A Nettles
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wendy Wenderski
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sam H Kim
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kyra C Roepke
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Claire Ellis
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hind Z Abuzaid
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gerald R Crabtree
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Kaiser J, Risteska A, Muller AG, Sun H, Lei B, Nay K, Means AR, Cousin MA, Drewry DH, Oakhill JS, Kemp BE, Hannan AJ, Berk M, Febbraio MA, Gundlach AL, Hill-Yardin EL, Scott JW. Convergence on CaMK4: A Key Modulator of Autism-Associated Signaling Pathways in Neurons. Biol Psychiatry 2025; 97:439-449. [PMID: 39442785 DOI: 10.1016/j.biopsych.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/03/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Although the precise underlying cause(s) of autism spectrum disorder remain unclear, more than 1000 rare genetic variations are associated with the condition. For many people living with profound autism, this genetic heterogeneity has impeded the identification of common biological targets for therapy development for core and comorbid traits that include significant impairments in social communication and repetitive and restricted behaviors. A substantial number of genes associated with autism encode proteins involved in signal transduction and synaptic transmission that are critical for brain development and function. CAMK4 is an emerging risk gene for autism spectrum disorder that encodes the CaMK4 (calcium/calmodulin-dependent protein kinase 4) enzyme. CaMK4 is a key component of a Ca2+-activated signaling pathway that regulates neurodevelopment and synaptic plasticity. In this review, we discuss 3 genetic variants of CAMK4 found in individuals with hyperkinetic movement disorder and comorbid neurological symptoms including autism spectrum disorder that are likely pathogenic with monogenic effect. We also comment on 4 other genetic variations in CAMK4 that show associations with autism spectrum disorder, as well as 12 examples of autism-associated variations in other genes that impact CaMK4 signaling pathways. Finally, we highlight 3 environmental risk factors that impact CaMK4 signaling based on studies of preclinical models of autism and/or clinical cohorts. Overall, we review molecular, genetic, physiological, and environmental evidence that suggest that defects in the CaMK4 signaling pathway may play an important role in a common autism pathogenesis network across numerous patient groups, and we propose CaMK4 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jacqueline Kaiser
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Alana Risteska
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Abbey G Muller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Haoxiong Sun
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Bethany Lei
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Kevin Nay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Margot A Cousin
- Center for Individualized Medicine, College of Medicine, Mayo Clinic, Rochester, Minnesota
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Berk
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Andrew L Gundlach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Elisa L Hill-Yardin
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia; School of Health and Biomedical Sciences, STEM College, RMIT University, Melbourne, Victoria, Australia.
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Liu L, Nguyen H, Das U, Ogunsola S, Yu J, Lei L, Kung M, Pejhan S, Rastegar M, Xie J. Epigenetic control of adaptive or homeostatic splicing during interval-training activities. Nucleic Acids Res 2024; 52:7211-7224. [PMID: 38661216 PMCID: PMC11229381 DOI: 10.1093/nar/gkae311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/03/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024] Open
Abstract
Interval-training activities induce adaptive cellular changes without altering their fundamental identity, but the precise underlying molecular mechanisms are not fully understood. In this study, we demonstrate that interval-training depolarization (ITD) of pituitary cells triggers distinct adaptive or homeostatic splicing responses of alternative exons. This occurs while preserving the steady-state expression of the Prolactin and other hormone genes. The nature of these splicing responses depends on the exon's DNA methylation status, the methyl-C-binding protein MeCP2 and its associated CA-rich motif-binding hnRNP L. Interestingly, the steady expression of the Prolactin gene is also reliant on MeCP2, whose disruption leads to exacerbated multi-exon aberrant splicing and overexpression of the hormone gene transcripts upon ITD, similar to the observed hyperprolactinemia or activity-dependent aberrant splicing in Rett Syndrome. Therefore, epigenetic control is crucial for both adaptive and homeostatic splicing and particularly the steady expression of the Prolactin hormone gene during ITD. Disruption in this regulation may have significant implications for the development of progressive diseases.
Collapse
Affiliation(s)
- Ling Liu
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Hai Nguyen
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Urmi Das
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Samuel Ogunsola
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jiankun Yu
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lei Lei
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Matthew Kung
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Shervin Pejhan
- Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mojgan Rastegar
- Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jiuyong Xie
- Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
4
|
Galaraga K, Rogaeva A, Biniam N, Daigle M, Albert PR. CaMKIV-Mediated Phosphorylation Inactivates Freud-1/CC2D1A Repression for Calcium-Dependent 5-HT1A Receptor Gene Induction. Int J Mol Sci 2024; 25:6194. [PMID: 38892382 PMCID: PMC11172825 DOI: 10.3390/ijms25116194] [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: 04/26/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Calcium calmodulin-dependent protein kinase (CaMK) mediates calcium-induced neural gene activation. CaMK also inhibits the non-syndromic intellectual disability gene, Freud-1/CC2D1A, a transcriptional repressor of human serotonin-1A (5-HT1A) and dopamine-D2 receptor genes. The altered expression of these Freud-1-regulated genes is implicated in mental illnesses such as major depression and schizophrenia. We hypothesized that Freud-1 is blocked by CaMK-induced phosphorylation. The incubation of purified Freud-1 with either CaMKIIα or CaMKIV increased Freud-1 phosphorylation that was partly prevented in Freud-1-Ser644Ala and Freud-1-Thr780Ala CaMK site mutants. In human SK-N-SH neuroblastoma cells, active CaMKIV induced the serine and threonine phosphorylation of Freud-1, and specifically increased Freud-1-Thr780 phosphorylation in transfected HEK-293 cells. The activation of purified CaMKIIα or CaMKIV reduced Freud-1 binding to its DNA element on the 5-HT1A and dopamine-D2 receptor genes. In SK-N-SH cells, active CaMKIV but not CaMKIIα blocked the Freud-1 repressor activity, while Freud-1 Ser644Ala, Thr780Ala or dual mutants were resistant to inhibition by activated CaMKIV or calcium mobilization. These results indicate that the Freud-1 repressor activity is blocked by CaMKIV-induced phosphorylation at Thr780, resulting in the up-regulation of the target genes, such as the 5-HT1A receptor gene. The CaMKIV-mediated inhibition of Freud-1 provides a novel de-repression mechanism to induce 5-HT1A receptor expression for the regulation of cognitive development, behavior and antidepressant response.
Collapse
Affiliation(s)
| | | | | | | | - Paul R. Albert
- Ottawa Hospital Research Institute (Neuroscience), Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON K1H-8M5, Canada; (K.G.); (A.R.); (N.B.); (M.D.)
| |
Collapse
|
5
|
Lu H, Jiang J, Min J, Huang X, McLeod P, Liu W, Haig A, Gunaratnam L, Jevnikar AM, Zhang ZX. The CaMK Family Differentially Promotes Necroptosis and Mouse Cardiac Graft Injury and Rejection. Int J Mol Sci 2024; 25:4428. [PMID: 38674016 PMCID: PMC11050252 DOI: 10.3390/ijms25084428] [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: 01/18/2024] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Organ transplantation is associated with various forms of programmed cell death which can accelerate transplant injury and rejection. Targeting cell death in donor organs may represent a novel strategy for preventing allograft injury. We have previously demonstrated that necroptosis plays a key role in promoting transplant injury. Recently, we have found that mitochondria function is linked to necroptosis. However, it remains unknown how necroptosis signaling pathways regulate mitochondrial function during necroptosis. In this study, we investigated the receptor-interacting protein kinase 3 (RIPK3) mediated mitochondrial dysfunction and necroptosis. We demonstrate that the calmodulin-dependent protein kinase (CaMK) family members CaMK1, 2, and 4 form a complex with RIPK3 in mouse cardiac endothelial cells, to promote trans-phosphorylation during necroptosis. CaMK1 and 4 directly activated the dynamin-related protein-1 (Drp1), while CaMK2 indirectly activated Drp1 via the phosphoglycerate mutase 5 (PGAM5). The inhibition of CaMKs restored mitochondrial function and effectively prevented endothelial cell death. CaMKs inhibition inhibited activation of CaMKs and Drp1, and cell death and heart tissue injury (n = 6/group, p < 0.01) in a murine model of cardiac transplantation. Importantly, the inhibition of CaMKs greatly prolonged heart graft survival (n = 8/group, p < 0.01). In conclusion, CaMK family members orchestrate cell death in two different pathways and may be potential therapeutic targets in preventing cell death and transplant injury.
Collapse
Affiliation(s)
- Haitao Lu
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
- Department of Pathology, Western University, London, ON N6A 3K7, Canada
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
| | - Jeffery Min
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
| | - Patrick McLeod
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
| | - Weihua Liu
- Department of Pathology, Western University, London, ON N6A 3K7, Canada
| | - Aaron Haig
- Department of Pathology, Western University, London, ON N6A 3K7, Canada
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
- Department of Microbiology and Immunology, Western University, London, ON N6A 3K7, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON N6A 5A5, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON N6A 3K7, Canada
| | - Anthony M. Jevnikar
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
- Department of Microbiology and Immunology, Western University, London, ON N6A 3K7, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON N6A 5A5, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON N6A 3K7, Canada
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON N6A 5A5, Canada; (H.L.); (A.M.J.)
- Department of Pathology, Western University, London, ON N6A 3K7, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON N6A 5A5, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
6
|
Gourisankar S, Wenderski W, Paulo JA, Kim SH, Roepke K, Ellis C, Gygi SP, Crabtree GR. Synaptic Activity Causes Minute-scale Changes in BAF Complex Composition and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562244. [PMID: 37873481 PMCID: PMC10592824 DOI: 10.1101/2023.10.13.562244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Genes encoding subunits of the SWI/SNF or BAF ATP-dependent chromatin remodeling complex are among the most enriched for deleterious de novo mutations in intellectual disabilities and autism spectrum disorder, but the causative molecular pathways are not fully known 1,2 . Synaptic activity in neurons is critical for learning and memory and proper neural development 3 . Neural activity prompts calcium influx and transcription within minutes, facilitated in the nucleus by various transcription factors (TFs) and chromatin modifiers 4 . While BAF is required for activity-dependent developmental processes such as dendritic outgrowth 5-7 , the immediate molecular consequences of neural activity on BAF complexes and their functions are unknown. Here we mapped minute-scale biochemical consequences of neural activity, modeled by membrane depolarization of embryonic mouse primary cortical neurons, on BAF complexes. We used acute chemical perturbations of BAF ATPase activity and kinase signaling to define the activity-dependent effects on BAF complexes and activity-dependent BAF functions. Our studies found that BAF complexes change in subunit composition and are selectively phosphorylated within 10 minutes of depolarization. Increased levels of the core PBAF subunit Baf200/ Arid2 , uniquely containing an RFX-like DNA-binding domain, are concurrent with ATPase-dependent opening of chromatin at RFX/X-box motifs. Changes in BAF composition and phosphorylation lead to the regulation of chromatin accessibility for critical neurogenesis TFs. These biochemical effects are a convergent phenomenon downstream of multiple growth factor signaling pathways in mouse neurons and fibroblasts suggesting that BAF integrates signaling information from the membrane. In support of such a membrane-to-nucleus signaling cascade, we also identified a BAF-interacting kinase, Dclk2, whose inhibition attenuates BAF phosphorylation selectively. Our findings support a direct role of BAF complexes in responding to synaptic activity to regulate TF binding and transcription.
Collapse
|
7
|
Zhao J, Dong L, Huo T, Cheng J, Li X, Huangfu X, Sun S, Wang H, Li L. O-GlcNAc Transferase (OGT) Protects Cerebral Neurons from Death During Ischemia/Reperfusion (I/R) Injury by Modulating Drp1 in Mice. Neuromolecular Med 2021; 24:299-310. [PMID: 34705256 DOI: 10.1007/s12017-021-08688-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 09/13/2021] [Indexed: 01/01/2023]
Abstract
Previous studies have demonstrated that increased O-linked N-acetylglucosamine (O-GlcNAc) level could promote cell survival following environmental stresses. This study aimed to explore the role of O-GlcNAc transferase (OGT) during cerebral ischemia/reperfusion (I/R) injury. The mouse model with cerebral I/R injury was induced by middle cerebral artery occlusion/reperfusion (MCAO/R). The expression of ogt in brain tissues was detected by qRT-PCR, Western blot, and immunohistochemistry (IHC) staining assay. Neurological deficit was evaluated using a modified scoring system. The infarct volume was assessed by TTC staining assay. Neuronal apoptosis in brain tissues was evaluated by TUNEL staining assay. The level of cleaved caspase-3 in brain tissues was detected by Western blot and IHC staining assay. The expression of critical proteins involved in mitochondrial fission, including OPA1, Mfn1, and Mfn2, as well as Mff and Drp1 was detected by Western blot and IHC, respectively. The expression of ogt during cerebral I/R injury was significantly upregulated. Ogt knockdown significantly increased neurological score and infarct volume in I/R-induced mice. Meanwhile, ogt knockdown significantly enhanced neuronal apoptosis and cleaved caspase-3 level in brain tissues of I/R-induced mice. In addition, ogt knockdown markedly decreased serine 637 phosphorylation level of mitochondrial fission protein dynamin-related protein 1 (Drp1) and promoted Drp1 translocation from the cytosol to the mitochondria. Moreover, the specific Drp1 inhibitor mdivi-1 effectively attenuated ogt knockdown-induced brain injury of I/R-stimulated mice in vivo. Our study revealed that OGT protects against cerebral I/R injury by inhibiting the function of Drp1 in mice, suggesting that ogt may be a potential therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Jingru Zhao
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Lipeng Dong
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Tiantian Huo
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Jinming Cheng
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Xiaojuan Li
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Xiaojuan Huangfu
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Sujuan Sun
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Hebo Wang
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China
| | - Litao Li
- Department of Neurology, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, P. R. China.
| |
Collapse
|
8
|
Takata T, Araki S, Tsuchiya Y, Watanabe Y. Persulfide Signaling in Stress-Initiated Calmodulin Kinase Response. Antioxid Redox Signal 2020; 33:1308-1319. [PMID: 32460522 DOI: 10.1089/ars.2020.8138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Significance: Calcium ion (Ca2+)/calmodulin (CaM)-dependent protein kinases (CaMKs) are activated by phosphorylation of a crucial threonine residue either by itself (CaMKII) or by upstream kinases, CaMK kinases (CaMKKs) (CaMKI and CaMKIV). CaMKs, present in most mammalian tissues, can phosphorylate many downstream targets, thereby regulating numerous cellular functions. Recent Advances: Aside from canonical post-translational modifications, cysteine-based redox switches in CaMKs affect their enzyme activities. In addition to reactive oxygen species (ROS) and reactive nitrogen species (RNS), reactive sulfur species (RSS) are also recognized as key signaling molecules, regulating protein function through polysulfidation, formation of polysulfides [-S-(S)n-H] on their reactive cysteine residues. To comprehend the biological significance of RSS signaling-related CaMK regulation, here we introduce a novel concept defining CaMKs as RSS targets in stress responses. The stress responses include an irreversible electrophile attack for CaMKI, inflammation for CaMKII, and endoplasmic reticulum stress for CaMKIV. Critical Issues: Development of various human diseases is associated with increased ROS, RNS, and RSS generation. Therefore, depending on specific pathophysiology, RSS could have very particular effects on CaMK functions. Future Directions: How multiple sources and mutual reactions of ROS, RNS, and RSS are coordinated is obscure. Elucidating the mechanisms through applications of enzymology, chemical biology, and mass spectrometry enables to uncover the complexities of redox regulation of CaMK cascades.
Collapse
Affiliation(s)
- Tsuyoshi Takata
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan.,Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoma Araki
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| | - Yukihiro Tsuchiya
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
9
|
Zech M, Bardakjian TM, Stoklosa M, Ploski R, Jech R, Gonzalez-Alegre P, Winkelmann J. A Neurodevelopmental Disorder With Dystonia and Chorea Resulting From Clustering CAMK4 Variants. Mov Disord 2020; 36:520-521. [PMID: 33211350 DOI: 10.1002/mds.28398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany.,Institute of Human Genetics, Technical University of Munich, Munich, Germany
| | - Tanya M Bardakjian
- Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Rafal Ploski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Robert Jech
- Department of Neurology, Charles University, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Pedro Gonzalez-Alegre
- Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany.,Institute of Human Genetics, Technical University of Munich, Munich, Germany.,Lehrstuhl für Neurogenetik, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
| |
Collapse
|
10
|
Cao K, Wang H, Fang Y, Wang Y, Wei L, Chen X, Jiang Z, Wei X, Hu Y. Histone Deacetylase 4 Promotes Osteosarcoma Cell Proliferation and Invasion by Regulating Expression of Proliferating Cell Nuclear Antigen. Front Oncol 2019; 9:870. [PMID: 31552187 PMCID: PMC6743440 DOI: 10.3389/fonc.2019.00870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/21/2019] [Indexed: 01/01/2023] Open
Abstract
Background/Aims: Osteosarcoma (OS) is commonly characterized by lower survival rates and high incidences of local recurrence due to its highly aggressive nature and metastatic tendencies. Studies have shown that histone deacetylase 4 (HDAC4) and proliferating cell nuclear antigen (PCNA) are highly expressed in cancers. Nevertheless, the roles of HDAC4 and PCNA in osteosarcoma (OS) remain unclear. This research aimed to study the expression of HDAC4 and PCNA and their relation to cell proliferation and invasion in human OS. Methods: The levels of HDAC4 and PCNA mRNA and protein were tested in human OS and osteochondroma (OC) tissues. The overexpression and knockdown of HDAC4 in OS cell lines were used to determine the effect of HDAC4 on the expression and degradation of PCNA. The effect of HDAC4 on cell proliferation, invasion and apoptosis was also detected. Additionally, we explored the interaction between HDAC4 and PCNA. Results: The results showed that both HDAC4 and PCNA were increased in human OS tissues. Overexpression of the HDAC4 protein increased the protein level of PCNA, had no effect on the PCNA mRNA level, and decreased the level of ubiquitinated PCNA. We found that overexpression of HDAC4 promoted cell proliferation and invasion and inhibited apoptosis. The opposite effects were observed when HDAC4 was knocked down. The results also showed that HDAC4 could bind to PCNA directly. Conclusions: Our findings suggest that HDAC4 could promote OS cell proliferation and invasion by regulating the expression of PCNA. Thus, our research indicates that HDAC4 may be a potential target for therapy in OS.
Collapse
Affiliation(s)
- Kun Cao
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Wang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yueyang Fang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuan Wang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Wei
- Department of Orthopaedics and Department of Surgery, Warren Alpert Medical School of Brown University/Rhode Island Hospital (RIH), Providence, RI, United States
| | - Xi Chen
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zheng Jiang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yong Hu
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Li X, Wang R, Zhang J, Yang S, Ji K, Du B, Liu X, Liu B, Qi S, Jia Q, Fan R. Cyclin-dependent kinase 5 regulates proliferation, migration, tyrosinase activity, and melanin production in B16-F10 melanoma cells via the essential regulator p-CREB. In Vitro Cell Dev Biol Anim 2019; 55:416-425. [PMID: 31069610 DOI: 10.1007/s11626-019-00343-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/22/2019] [Indexed: 01/18/2023]
Abstract
Melanoma is an aggressive cancer with increasing incidence and a growing lifetime risk that arises from normal melanocytes or their precursors. A thorough understanding of the molecular mechanism of melanomagenesis and melanoma biology is essential for the diagnosis, prognostication, and therapy of melanoma. Cyclin-dependent protein kinase 5 (Cdk5) is one of the proteins highly expressed in B16-F10 melanoma cells that controls melanoma cell motility, invasiveness, and metastatic spread and might be a promising novel therapeutic target. The effect of Cdk5 on proliferation and migration, which are important for carcinogenesis, has not been reported. In the current study, we found that siRNA-mediated knockdown of Cdk5 in B16-F10 melanoma cells inhibited melanoma cell proliferation through downregulation of the CaMK4-p-CREB pathway, inhibited migration through downregulation of p-CREB, integrin beta 1, and integrin beta 5, and also inhibited tyrosinase activity and melanin production through p-CREB-MITF regulation. The results indicate that Cdk5 controls melanoma development, with an essential regulatory role for p-CREB.
Collapse
Affiliation(s)
- Xiuqing Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Ruifang Wang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Junzhen Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Shanshan Yang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Kaiyuan Ji
- College of Animal Science and Veterinary Medicine, Anhui Agricultural University, Changjiang West Road, Hefei, 230036, China
| | - Bin Du
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Xuexian Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Bo Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Shuhui Qi
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Qiong Jia
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China
| | - Ruiwen Fan
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Mingxian South Road, Taigu, 030801, China.
| |
Collapse
|
12
|
Brzozowski JS, Skelding KA. The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention. Pharmaceuticals (Basel) 2019; 12:ph12010008. [PMID: 30621060 PMCID: PMC6469190 DOI: 10.3390/ph12010008] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 01/25/2023] Open
Abstract
The importance of Ca2+ signalling in key events of cancer cell function and tumour progression, such as proliferation, migration, invasion and survival, has recently begun to be appreciated. Many cellular Ca2+-stimulated signalling cascades utilise the intermediate, calmodulin (CaM). The Ca2+/CaM complex binds and activates a variety of enzymes, including members of the multifunctional Ca2+/calmodulin-stimulated protein kinase (CaMK) family. These enzymes control a broad range of cancer-related functions in a multitude of tumour types. Herein, we explore the cancer-related functions of these kinases and discuss their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joshua S Brzozowski
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Kathryn A Skelding
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
13
|
Zech M, Lam DD, Weber S, Berutti R, Poláková K, Havránková P, Fečíková A, Strom TM, Růžička E, Jech R, Winkelmann J. A unique de novo gain-of-function variant in CAMK4 associated with intellectual disability and hyperkinetic movement disorder. Cold Spring Harb Mol Case Stud 2018; 4:mcs.a003293. [PMID: 30262571 PMCID: PMC6318768 DOI: 10.1101/mcs.a003293] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/04/2018] [Indexed: 01/12/2023] Open
Abstract
Calcium/calmodulin-dependent protein kinases (CaMKs) are key mediators of calcium signaling and underpin neuronal health. Although widely studied, the contribution of CaMKs to Mendelian disease is rather enigmatic. Here, we describe an unusual neurodevelopmental phenotype, characterized by milestone delay, intellectual disability, autism, ataxia, and mixed hyperkinetic movement disorder including severe generalized dystonia, in a proband who remained etiologically undiagnosed despite exhaustive testing. We performed trio whole-exome sequencing to identify a de novo essential splice-site variant (c.981+1G>A) in CAMK4, encoding CaMKIV. Through in silico evaluation and cDNA analyses, we demonstrated that c.981+1G>A alters CAMK4 pre-mRNA processing and results in a stable mRNA transcript containing a 77-nt out-of-frame deletion and a premature termination codon within the last exon. The expected protein, p.Lys303Serfs*28, exhibits selective loss of the carboxy-terminal regulatory domain of CaMKIV and bears striking structural resemblance to previously reported synthetic mutants that confer constitutive CaMKIV activity. Biochemical studies in proband-derived cells confirmed an activating effect of c.981+1G>A and indicated that variant-induced excessive CaMKIV signaling is sensitive to pharmacological manipulation. Additionally, we found that variants predicted to cause selective depletion of CaMKIV's regulatory domain are unobserved in diverse catalogs of human variation, thus revealing that c.981+1G>A is a unique molecular event. We propose that our proband's phenotype is explainable by a dominant CAMK4 splice-disrupting mutation that acts through a gain-of-function mechanism. Our findings highlight the importance of CAMK4 in human neurodevelopment, provide a foundation for future clinical research of CAMK4, and suggest the CaMKIV signaling pathway as a potential drug target in neurological disease.
Collapse
Affiliation(s)
- Michael Zech
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany.,Klinik und Poliklinik für Neurologie, Klinikum rechts der Isar, Technische Universität München, Munich, 81675, Germany
| | - Daniel D Lam
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Sandrina Weber
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Riccardo Berutti
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Kamila Poláková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Petra Havránková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Anna Fečíková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Tim M Strom
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, 85764, Germany.,Institut für Humangenetik, Technische Universität München, Munich, 81675, Germany
| | - Evžen Růžička
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Robert Jech
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague, 120 00, Czech Republic
| | - Juliane Winkelmann
- Institut für Neurogenomik, Helmholtz Zentrum München, Munich, 85764, Germany.,Institut für Humangenetik, Technische Universität München, Munich, 81675, Germany.,Lehrstuhl für Neurogenetik, Technische Universität München, Munich, 80333, Germany.,Munich Cluster for Systems Neurology, SyNergy, Munich, 81377, Germany
| |
Collapse
|
14
|
Laarse SAM, Leney AC, Heck AJR. Crosstalk between phosphorylation and O‐Glc
NA
cylation: friend or foe. FEBS J 2018; 285:3152-3167. [DOI: 10.1111/febs.14491] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/27/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Saar A. M. Laarse
- Biomolecular Mass Spectrometry and Proteomics Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences Utrecht University The Netherlands
- Netherlands Proteomics Centre Utrecht The Netherlands
| | - Aneika C. Leney
- Biomolecular Mass Spectrometry and Proteomics Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences Utrecht University The Netherlands
- Netherlands Proteomics Centre Utrecht The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences Utrecht University The Netherlands
- Netherlands Proteomics Centre Utrecht The Netherlands
| |
Collapse
|
15
|
Takemoto-Kimura S, Suzuki K, Horigane SI, Kamijo S, Inoue M, Sakamoto M, Fujii H, Bito H. Calmodulin kinases: essential regulators in health and disease. J Neurochem 2017; 141:808-818. [PMID: 28295333 DOI: 10.1111/jnc.14020] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/08/2017] [Indexed: 01/22/2023]
Abstract
Neuronal activity induces intracellular Ca2+ increase, which triggers activation of a series of Ca2+ -dependent signaling cascades. Among these, the multifunctional Ca2+ /calmodulin-dependent protein kinases (CaMKs, or calmodulin kinases) play key roles in neuronal transmission, synaptic plasticity, circuit development and cognition. The most investigated CaMKs for these roles in neuronal functions are CaMKI, CaMKII, CaMKIV and we will shed light on these neuronal CaMKs' functions in this review. Catalytically active members of CaMKs currently are CaMKI, CaMKII, CaMKIV and CaMKK. Although they all necessitate the binding of Ca2+ and calmodulin complex (Ca2+ /CaM) for releasing autoinhibition, each member of CaMK has distinct activation mechanisms-autophosphorylation mediated autonomy of multimeric CaMKII and CaMKK-dependent phosphoswitch-induced activation of CaMKI or CaMKIV. Furthermore, each CaMK shows distinct subcellular localization that underlies specific compartmentalized function in each activated neuron. In this review, we first summarize these molecular characteristics of each CaMK as to regulation and subcellular localization, and then describe each biological function. In the last section, we also focus on the emerging role of CaMKs in pathophysiological conditions by introducing the recent studies, especially focusing on drug addiction and depression, and discuss how dysfunctional CaMKs may contribute to the pathology of the neuropsychological disorders. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Japan.,PRESTO-Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Kanzo Suzuki
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Japan
| | - Satoshi Kamijo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Inoue
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masayuki Sakamoto
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Naz H, Shahbaaz M, Bisetty K, Islam A, Ahmad F, Hassan MI. Effect of pH on the structure, function, and stability of human calcium/calmodulin-dependent protein kinase IV: combined spectroscopic and MD simulation studies. Biochem Cell Biol 2016; 94:221-8. [PMID: 27032767 DOI: 10.1139/bcb-2015-0132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human calcium/calmodulin-dependent protein kinase IV (CAMKIV) is a member of Ser/Thr protein kinase family. It is regulated by the calcium-calmodulin dependent signal through a secondary messenger, Ca(2+), which leads to the activation of its autoinhibited form. The over-expression and mutation in CAMKIV as well as change in Ca(2+) concentration is often associated with numerous neurodegenerative diseases and cancers. We have successfully cloned, expressed, and purified a functionally active kinase domain of human CAMKIV. To observe the effect of different pH conditions on the structural and functional properties of CAMKIV, we have used spectroscopic techniques such as circular diachroism (CD) absorbance and fluorescence. We have observed that within the pH range 5.0-11.5, CAMKIV maintained both its secondary and tertiary structures, along with its function, whereas significant aggregation was observed at acidic pH (2.0-4.5). We have also performed ATPase activity assays under different pH conditions and found a significant correlation between the structure and enzymatic activities of CAMKIV. In-silico validations were further carried out by modeling the 3-dimensional structure of CAMKIV and then subjecting it to molecular dynamics (MD) simulations to understand its conformational behavior in explicit water conditions. A strong correlation between spectroscopic observations and the output of molecular dynamics simulation was observed for CAMKIV.
Collapse
Affiliation(s)
- Huma Naz
- a Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohd Shahbaaz
- b Department of Chemistry, Durban University of Technology, Durban-4000, South Africa
| | - Krishna Bisetty
- b Department of Chemistry, Durban University of Technology, Durban-4000, South Africa
| | - Asimul Islam
- a Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Faizan Ahmad
- a Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- a Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| |
Collapse
|
17
|
Calcium/calmodulin-dependent protein kinase IV: A multifunctional enzyme and potential therapeutic target. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:54-65. [PMID: 26773169 DOI: 10.1016/j.pbiomolbio.2015.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/21/2015] [Accepted: 12/28/2015] [Indexed: 01/23/2023]
Abstract
The calcium/calmodulin-dependent protein kinase IV (CAMKIV) belongs to the serine/threonine protein kinase family, and is primarily involved in transcriptional regulation in lymphocytes, neurons and male germ cells. CAMKIV operates the signaling cascade and regulates activity of several transcription activators by phosphorylation, which in turn plays pivotal roles in immune response, inflammation and memory consolidation. In this review, we tried to focus on different aspects of CAMKIV to understand the significance of this protein in the biological system. This enzyme is associated with varieties of disorders such as cerebral hypoxia, azoospermia, endometrial and ovarian cancer, systemic lupus, etc., and hence it is considered as a potential therapeutic target. Structure of CAMKIV is comprised of five distinct domains in which kinase domain is responsible for enzyme activity. CAMKIV is involved in varieties of cellular functions such as regulation of gene expression, T-cell maturation, regulation of survival phase of dendritic cells, bone growth and metabolism, memory consolidation, sperm motility, regulation of microtubule dynamics, cell-cycle progression and apoptosis. In this review, we performed an extensive analysis on structure, function and regulation of CAMKIV and associated diseases.
Collapse
|
18
|
Cao K, Wei L, Zhang Z, Guo L, Zhang C, Li Y, Sun C, Sun X, Wang S, Li P, Wei X. Decreased histone deacetylase 4 is associated with human osteoarthritis cartilage degeneration by releasing histone deacetylase 4 inhibition of runt-related transcription factor-2 and increasing osteoarthritis-related genes: a novel mechanism of human osteoarthritis cartilage degeneration. Arthritis Res Ther 2014; 16:491. [PMID: 25424126 PMCID: PMC4265470 DOI: 10.1186/s13075-014-0491-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 11/12/2014] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION To investigate if decreased histone deacetylase 4 (HDAC4) is associated with human osteoarthritis (OA) cartilage degeneration by releasing HDAC4 inhibition of runt-related transcription factor-2 (Runx2) resulting in increase of OA cartilage degeneration-related genes. METHODS The mRNA and protein levels of HDAC4, Runx2, matrix metalloproteinase (MMP)-13, Indian hedgehog (Ihh) and type X collagen were detected by performing real-time PCR (RT-PCR), western blotting and immunohistochemistry on specimens from human OA and normal cartilage. To further explore the mechanism of regulation of Runx2 and OA-related genes by HDAC4, changes in these OA-related genes were further quantified by RT-PCR after overexpression of HDAC4 and knockdown of HDAC4 by siRNA. Runx2 and MMP-13 promoter activities were measured by dual luciferase assays. RESULTS The levels of HDAC4 in the cartilage from OA patients and healthy 40- to 60-year-old donors were decreased to 31% and 65% compared with specimens from 20- to 40-year-old healthy donors, respectively (P <0.05). Decreased HDAC4 was associated with increased Runx2 and other OA-related genes in human OA cartilage, specifically: MMP-13, Ihh and type X collagen. Exogenous HDAC4 decreased the mRNA levels of Runx2, MMP1, MMP3, MMP-13, type X collagen, Ihh, ADAMTS-4 and -5, and increased the mRNA of type II collagen. In addition, the data also shows that overexpression of HDAC4 not only decreased the expression of interleukin (IL)-1β, Cox2 and iNos and increased the expression of aggrecan, but also partially blocked the effect of IL-1β on expression of catabolic events in human OA chondrocytes. HDAC4 also inhibited Runx2 promoter activity and MMP13 promotor activity in a dose-dependent manner. In contrast, inhibition of HDAC4 by TSA drug had an opposite effect. CONCLUSIONS Our study is the first to demonstrate that decreased HDAC4 contributes, at least in part, to the pathogenesis of OA cartilage degeneration. Thus, HDAC4 may have chondroprotective properties by inhibiting Runx2 and OA-related genes.
Collapse
|
19
|
Razanau A, Xie J. Emerging mechanisms and consequences of calcium regulation of alternative splicing in neurons and endocrine cells. Cell Mol Life Sci 2013; 70:4527-36. [PMID: 23800988 PMCID: PMC11113957 DOI: 10.1007/s00018-013-1390-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/21/2013] [Accepted: 05/27/2013] [Indexed: 12/12/2022]
Abstract
Alternative splicing contributes greatly to proteomic complexity. How it is regulated by external stimuli to sculpt cellular properties, particularly the highly diverse and malleable neuronal properties, is an underdeveloped area of emerging interest. A number of recent studies in neurons and endocrine cells have begun to shed light on its regulation by calcium signals. Some mechanisms include changes in the trans-acting splicing factors by phosphorylation, protein level, alternative pre-mRNA splicing, and nucleocytoplasmic redistribution of proteins to alter protein-RNA or protein-protein interactions, as well as modulation of chromatin states. Importantly, functional analyses of the control of specific exons/splicing factors in the brain point to a crucial role of this regulation in synaptic maturation, maintenance, and transmission. Furthermore, its deregulation has been implicated in the pathogenesis of neurological disorders, particularly epilepsy/seizure. Together, these studies have not only provided mechanistic insights into the regulation of alternative splicing by calcium signaling but also demonstrated its impact on neuron differentiation, function, and disease. This may also help our understanding of similar regulations in other types of cells.
Collapse
Affiliation(s)
- Aleh Razanau
- Department of Physiology, University of Manitoba, 439 BMSB, 745 Bannatyne Ave, Winnipeg, R3E 0J9 Canada
| | - Jiuyong Xie
- Department of Physiology, University of Manitoba, 439 BMSB, 745 Bannatyne Ave, Winnipeg, R3E 0J9 Canada
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9 Canada
| |
Collapse
|
20
|
Guo L, Stripay JL, Zhang X, Collage RD, Hulver M, Carchman EH, Howell GM, Zuckerbraun BS, Lee JS, Rosengart MR. CaMKIα regulates AMP kinase-dependent, TORC-1-independent autophagy during lipopolysaccharide-induced acute lung neutrophilic inflammation. THE JOURNAL OF IMMUNOLOGY 2013; 190:3620-8. [PMID: 23447692 DOI: 10.4049/jimmunol.1102975] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Autophagy is an evolutionarily conserved cytoplasmic process regulated by the energy rheostats mammalian target of rapamycin and AMP kinase (AMPK) that recycles damaged or unused proteins and organelles. It has been described as an important effector arm of immune cells. We have shown that the cytoplasmically oriented calcium/calmodulin-dependent protein kinase (CaMK)Iα regulates the inflammatory phenotype of the macrophage (M). In this study, we hypothesize that CaMKIα mediates M autophagy. LPS induced autophagy in RAW 264.7 cells and murine peritoneal M that was attenuated with biochemical CaMK inhibition or CaMKIα small interfering RNA (siRNA). Inhibition of CaMKIα reduced LPS-induced p-Thr(172)AMPK and target of rapamycin complex-1 activity, and expression of a constitutively active CaMKIα but not a kinase-deficient mutant induced p-Thr(172)AMPK and autophagy that was attenuated by the AMPK inhibitor compound C. Coimmunoprecipitation and in vitro kinase assays demonstrated that CaMKIα activates AMPK, thereby inducing ATG7, which also localizes to this CaMKIα/AMPK complex. During LPS-induced lung inflammation, C57BL/6 mice receiving CaMKIα(siRNA) displayed reduced lung and bronchoalveolar immune cell autophagy that correlated with reduced neutrophil recruitment, myeloperoxidase activity, and air space cytokine concentration. Independently inhibiting autophagy, using siRNA targeting the PI3K VPS34, yielded similar reductions in lung autophagy and neutrophil recruitment. Thus, a novel CaMKIα/AMPK pathway is rapidly activated in M exposed to LPS and regulates an early autophagic response, independent of target of rapamycin complex-1 inhibition. These mechanisms appear to be operant in vivo in orchestrating LPS-induced lung neutrophil recruitment and inflammation.
Collapse
Affiliation(s)
- Lanping Guo
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Liu B, Barbosa-Sampaio H, Jones PM, Persaud SJ, Muller DS. The CaMK4/CREB/IRS-2 cascade stimulates proliferation and inhibits apoptosis of β-cells. PLoS One 2012; 7:e45711. [PMID: 23049845 PMCID: PMC3458088 DOI: 10.1371/journal.pone.0045711] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/23/2012] [Indexed: 01/09/2023] Open
Abstract
Progressive reduction in β-cell mass is responsible for the development of type 2 diabetes mellitus, and alteration in insulin receptor substrate 2 (IRS-2) abundance plays a critical role in this process. IRS-2 expression is stimulated by the transcription factor cAMP response element-binding protein (CREB) and we recently demonstrated that Ca2+/calmodulin dependent kinase 4 (CaMK4) is upstream of CREB activation in β-cells. This study investigated whether CaMK4 is also a potential target to increase β-cell mass through CREB-mediated IRS-2 expression, by quantifying mouse MIN6 β-cell proliferation and apoptosis following IRS-2 knockdown, CaMKs inhibition and alterations in CaMK4 and CREB expression. Expression of constitutively active CaMK4 (ΔCaMK4) and CREB (CREBDIEDLM) significantly stimulated β-cell proliferation and survival. In contrast, expression of their corresponding dominant negative forms (ΔK75ECaMK4 and CREBM1) and silencing of IRS-2 increased apoptosis and reduced β-cell division. Moreover, CREBDIEDLM and CREBM1 expression completely abolished the effects of ΔK75ECaMK4 and of ΔCaMK4, respectively. Our results indicate that CaMK4 regulates β-cell proliferation and apoptosis in a CREB-dependent manner and that CaMK4-induced IRS-2 expression is important in these processes.
Collapse
Affiliation(s)
- Bo Liu
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| | - Helena Barbosa-Sampaio
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| | - Peter M. Jones
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| | - Shanta J. Persaud
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
- * E-mail:
| | - Dany S. Muller
- Diabetes Research Group, School of Medicine, Division of Diabetes & Nutritional Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
22
|
Ojuka EO, Goyaram V, Smith JAH. The role of CaMKII in regulating GLUT4 expression in skeletal muscle. Am J Physiol Endocrinol Metab 2012; 303:E322-31. [PMID: 22496345 DOI: 10.1152/ajpendo.00091.2012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Contractile activity during physical exercise induces an increase in GLUT4 expression in skeletal muscle, helping to improve glucose transport capacity and insulin sensitivity. An important mechanism by which exercise upregulates GLUT4 is through the activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in response to elevated levels of cytosolic Ca(2+) during muscle contraction. This review discusses the mechanism by which Ca(2+) activates CaMKII, explains research techniques currently used to alter CaMK activity in cells, and highlights various exercise models and pharmacological agents that have been used to provide evidence that CaMKII plays an important role in regulating GLUT4 expression. With regard to transcriptional mechanisms, the key research studies that identified myocyte enhancer factor 2 (MEF2) and GLUT4 enhancer factor as the major transcription factors regulating glut4 gene expression, together with their binding domains, are underlined. Experimental evidence showing that CaMK activation induces hyperacetylation of histones in the vicinity of the MEF2 domain and increases MEF2 binding to its cis element to influence MEF2-dependent Glut4 gene expression are also given along with data suggesting that p300 might be involved in acetylating histones on the Glut4 gene. Finally, an appraisal of the roles of other calcium- and non-calcium-dependent mechanisms, including the major HDAC kinases in GLUT4 expression, is also given.
Collapse
Affiliation(s)
- Edward O Ojuka
- University of Capetown/Medical Research Center Research Unit for Exercise Science & Sports Medicine, Department of Human Biology, Univeristy of Cape Town, Cape Town, South Africa.
| | | | | |
Collapse
|
23
|
Guan Y, Chen Q, Yang X, Haines P, Pei M, Terek R, Wei X, Zhao T, Wei L. Subcellular relocation of histone deacetylase 4 regulates growth plate chondrocyte differentiation through Ca2+/calmodulin-dependent kinase IV. Am J Physiol Cell Physiol 2012; 303:C33-40. [PMID: 22442139 DOI: 10.1152/ajpcell.00348.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regulatory mechanisms of chondrocyte differentiation in the growth plate are incompletely understood. Here, we find that histone deacetylase 4 (HDAC4) is located in the nucleus of chondrocytes in the proliferation zone and relocates to the cytoplasm of chondrocytes in the prehypertrophic zone in vivo. This suggests that the relocation of HDAC4 from the nucleus to the cytoplasm may play a role during chondrocyte differentiation. Expression of active CaMKIV in chondrocytes promotes HDAC4 relocation into cytoplasm in primary chondrocytes. Conversely, HDAC4 relocation is blocked by a Ca(2+)/calmodulin-dependent kinase IV (CaMKIV) inhibitor. This indicates that CaMKIV signaling plays an important role in regulating HDAC4 relocation. In addition, CaMKIV is required for HDAC4 phosphorylation, which is required for HDAC4 association with the cytoplasmic protein 14-3-3. Active CaMKIV also stimulates runt-related transcription factor-2 (RunX2) and type X collagen (Col X) promoter activities and overcomes repression of these promoter activities by HDAC4. Furthermore, CaMKIV increases gene expression of the chondrocyte differentiation markers Ihh and Col X. Our results demonstrate that CaMKIV induces chondrocyte differentiation through regulation of HDAC4 subcellular relocation, from the nucleus to the cytoplasm, which results in increased activity of RunX2 and transition of chondrocytes from the proliferative to the prehypertrophic stage. Thus, CaMKIV plays an important regulatory role during chondrocyte differentiation.
Collapse
Affiliation(s)
- Yingjie Guan
- Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Green MF, Scott JW, Steel R, Oakhill JS, Kemp BE, Means AR. Ca2+/Calmodulin-dependent protein kinase kinase beta is regulated by multisite phosphorylation. J Biol Chem 2011; 286:28066-79. [PMID: 21669867 PMCID: PMC3151052 DOI: 10.1074/jbc.m111.251504] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/01/2011] [Indexed: 01/04/2023] Open
Abstract
Ca(2+)/calmodulin-dependent protein kinase kinase β (CaMKKβ) is a serine/threonine-directed kinase that is activated following increases in intracellular Ca(2+). CaMKKβ activates Ca(2+)/calmodulin-dependent protein kinase I, Ca(2+)/calmodulin-dependent protein kinase IV, and the AMP-dependent protein kinase in a number of physiological pathways, including learning and memory formation, neuronal differentiation, and regulation of energy balance. Here, we report the novel regulation of CaMKKβ activity by multisite phosphorylation. We identify three phosphorylation sites in the N terminus of CaMKKβ, which regulate its Ca(2+)/calmodulin-independent autonomous activity. We then identify the kinases responsible for these phosphorylations as cyclin-dependent kinase 5 (CDK5) and glycogen synthase kinase 3 (GSK3). In addition to regulation of autonomous activity, we find that phosphorylation of CaMKKβ regulates its half-life. We find that cellular levels of CaMKKβ correlate with CDK5 activity and are regulated developmentally in neurons. Finally, we demonstrate that appropriate phosphorylation of CaMKKβ is critical for its role in neurite development. These results reveal a novel regulatory mechanism for CaMKKβ-dependent signaling cascades.
Collapse
Affiliation(s)
- Michelle F. Green
- From the Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710 and
| | - John W. Scott
- the Protein Chemistry and Metabolism Unit, St. Vincent's Institute of Medical Research, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Rohan Steel
- the Protein Chemistry and Metabolism Unit, St. Vincent's Institute of Medical Research, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Jonathan S. Oakhill
- the Protein Chemistry and Metabolism Unit, St. Vincent's Institute of Medical Research, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Bruce E. Kemp
- the Protein Chemistry and Metabolism Unit, St. Vincent's Institute of Medical Research, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Anthony R. Means
- From the Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710 and
| |
Collapse
|
25
|
Zhang X, Guo L, Collage RD, Stripay JL, Tsung A, Lee JS, Rosengart MR. Calcium/calmodulin-dependent protein kinase (CaMK) Ialpha mediates the macrophage inflammatory response to sepsis. J Leukoc Biol 2011; 90:249-61. [PMID: 21372190 PMCID: PMC3133437 DOI: 10.1189/jlb.0510286] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 12/31/2010] [Accepted: 02/01/2011] [Indexed: 11/24/2022] Open
Abstract
Dysregulated Ca(2+) handling is prevalent during sepsis and postulated to perpetuate the aberrant inflammation underlying subsequent organ dysfunction and death. The signal transduction cascades mediating these processes are unknown. Here, we identify that CaMKIα mediates the Mϕ response to LPS in vitro and the inflammation and organ dysfunction of sepsis in vivo. We show that LPS induced active pThr(177)-CaMKIα in RAW 264.7 cells and murine peritoneal Mϕ, which if inhibited biochemically with STO609 (CaMKK inhibitor) or by RNAi, reduces LPS-induced production of IL-10. Transfection of constitutively active CaMKIα (CaMKI293), but not a kinase-deficient mutant (CaMKI293(K49A)), induces IL-10 release. This production of IL-10 is mediated by CaMKIα-dependent regulation of p38 MAPK activation. CaMKIα activity also mediates the cellular release of HMGB1 by colocalizing with and regulating the packaging of HMGB1 into secretory lysosomes. During endotoxemia, mice receiving in vivo CaMKIα(RNAi) display reduced systemic concentrations of IL-10 and HMGB1 in comparison with mice receiving NT(RNAi). These data support the biological relevance of CaMKIα-dependent IL-10 production and HMGB1 secretion. In a CLP model of sepsis, CaMKIα(RNAi) mice display reduced systemic concentrations of IL-10, IL-6, TNF-α, and HMGB1 in comparison with NT(RNAi) mice, which correlate with reductions in the development of renal dysfunction. These data support that CaMKIα signaling is integral to the Mϕ responding to LPS and may also be operant in vivo in regulating the inflammation and organ dysfunction consequent to sepsis.
Collapse
Affiliation(s)
| | | | | | | | | | - Janet S. Lee
- Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
26
|
Wayman GA, Tokumitsu H, Davare MA, Soderling TR. Analysis of CaM-kinase signaling in cells. Cell Calcium 2011; 50:1-8. [PMID: 21529938 PMCID: PMC3236032 DOI: 10.1016/j.ceca.2011.02.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/15/2011] [Accepted: 02/17/2011] [Indexed: 12/18/2022]
Abstract
A change in intracellular free calcium is a common signaling mechanism that modulates a wide array of physiological processes in most cells. Responses to increased intracellular Ca(2+) are often mediated by the ubiquitous protein calmodulin (CaM) that upon binding Ca(2+) can interact with and alter the functionality of numerous proteins including a family of protein kinases referred to as CaM-kinases (CaMKs). Of particular interest are multifunctional CaMKs, such as CaMKI, CaMKII, CaMKIV and CaMKK, that can phosphorylate multiple downstream targets. This review will outline several protocols we have used to identify which members and/or isoforms of this CaMK family mediate specific cellular responses with a focus on studies in neurons. Many previous studies have relied on a single approach such as pharmacological inhibitors or transfected dominant-negative kinase constructs. Since each of these protocols has its limitations, that will be discussed, we emphasize the necessity to use multiple, independent approaches in mapping out cellular signaling pathways.
Collapse
Affiliation(s)
- Gary A Wayman
- Program in Neuroscience, Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University, Pullman, WA, USA.
| | | | | | | |
Collapse
|
27
|
Nimura T, Sugiyama Y, Sueyoshi N, Shigeri Y, Ishida A, Kameshita I. A minimum size homologue of Ca2+/calmodulin-dependent protein kinase IV naturally occurring in zebrafish. J Biochem 2010; 147:857-65. [DOI: 10.1093/jb/mvq021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
28
|
Ramakrishnan R, Dow EC, Rice AP. Characterization of Cdk9 T-loop phosphorylation in resting and activated CD4(+) T lymphocytes. J Leukoc Biol 2009; 86:1345-50. [PMID: 19741158 DOI: 10.1189/jlb.0509309] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The cellular kinase complex P-TEFb is composed of Cdk9 and cyclin T, and it is required for expression of most protein-coding genes by RNAP II. Cdk9 has been shown recently to be activated in cis by autophosphorylation of Thr186 in its T-loop. Using a phosphospecific Cdk9 antibody, we examined the level of Cdk9 T-loop phosphorylation in resting and activated CD4(+) T lymphocytes. Cdk9 T-loop phosphorylation was found to be low-to-undetectable in resting CD4(+) T lymphocytes, and upon activation by distinct stimuli, there is a rapid (<1 h) increase in pCdk9 that does not require protein synthesis. The low level of Cdk9 T-loop phosphorylation was not to be a result of the absence of an associated regulatory cyclin partner. These observations suggest that autophosphorylation of the Cdk9 T-loop is repressed in resting CD4(+) T lymphocytes. The low level of T-loop phosphorylation in resting cells is also reflected in a low level of phosphorylation of Ser2 in the carboxyl terminal domain of RNAP II, suggesting that lack of Cdk9 T-loop autophosphorylation may limit RNAP II elongation in quiescent CD4(+) T lymphocytes.
Collapse
Affiliation(s)
- Rajesh Ramakrishnan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
29
|
Reece KM, Mazalouskas MD, Wadzinski BE. The Balpha and Bdelta regulatory subunits of PP2A are necessary for assembly of the CaMKIV.PP2A signaling complex. Biochem Biophys Res Commun 2009; 386:582-7. [PMID: 19538941 PMCID: PMC2745967 DOI: 10.1016/j.bbrc.2009.06.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 06/10/2009] [Indexed: 11/16/2022]
Abstract
Calcium/calmodulin-dependent protein kinase IV (CaMKIV) is a serine/threonine kinase that is important in synaptic plasticity and T cell maturation. Activation of CaMKIV requires calcium/calmodulin binding and phosphorylation at T200 by CaMK kinase. Our previous work has shown that protein serine/threonine phosphatase 2A (PP2A) forms a complex with CaMKIV and negatively regulates its activity. Here we demonstrate that PP2A tightly regulates T200 phosphorylation of endogenous CaMKIV, but has little effect on the phosphorylation of the ectopically-expressed kinase. This differential regulation of endogenous versus exogenous CaMKIV is due to differences in their ability to associate with PP2A, as exogenous CaMKIV associates poorly with PP2A in comparison to endogenous CaMKIV. The inability of exogenous CaMKIV to associate with PP2A appears to be due to limiting amounts of endogenous PP2A regulatory B subunits, since coexpression of Balpha or Bdelta causes the recruitment of PP2Ac to ectopic CaMKIV, leading to formation of a CaMKIV.PP2A complex. Together, these data indicate that the B subunits are essential for the interaction of PP2A with CaMKIV.
Collapse
Affiliation(s)
- Kelie M Reece
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | |
Collapse
|
30
|
Takemura M, Mishima T, Wang Y, Kasahara J, Fukunaga K, Ohashi K, Mizuno K. Ca2+/calmodulin-dependent protein kinase IV-mediated LIM kinase activation is critical for calcium signal-induced neurite outgrowth. J Biol Chem 2009; 284:28554-62. [PMID: 19696021 DOI: 10.1074/jbc.m109.006296] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Actin cytoskeletal remodeling is essential for neurite outgrowth. LIM kinase 1 (LIMK1) regulates actin cytoskeletal remodeling by phosphorylating and inactivating cofilin, an actin filament-disassembling factor. In this study, we investigated the role of LIMK1 in calcium signal-induced neurite outgrowth. The calcium ionophore ionomycin induced LIMK1 activation and cofilin phosphorylation in Neuro-2a neuroblastoma cells. Knockdown of LIMK1 or expression of a kinase-dead mutant of LIMK1 suppressed ionomycin-induced cofilin phosphorylation and neurite outgrowth in Neuro-2a cells. Ionomycin-induced cofilin phosphorylation and neurite outgrowth were also blocked by KN-93, an inhibitor of Ca(2+)/calmodulin-dependent protein kinases (CaMKs), and STO-609, an inhibitor of CaMK kinase. An active form of CaMKIV but not CaMKI enhanced Thr-508 phosphorylation of LIMK1 and increased the kinase activity of LIMK1. Moreover, the active form of CaMKIV induced cofilin phosphorylation and neurite outgrowth, and a dominant negative form of CaMKIV suppressed ionomycin-induced neurite outgrowth. Taken together, our results suggest that LIMK1-mediated cofilin phosphorylation is critical for ionomycin-induced neurite outgrowth and that CaMKIV mediates ionomycin-induced LIMK1 activation.
Collapse
Affiliation(s)
- Miyohiko Takemura
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Dias WB, Cheung WD, Wang Z, Hart GW. Regulation of calcium/calmodulin-dependent kinase IV by O-GlcNAc modification. J Biol Chem 2009; 284:21327-37. [PMID: 19506079 PMCID: PMC2755857 DOI: 10.1074/jbc.m109.007310] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 05/22/2009] [Indexed: 12/21/2022] Open
Abstract
Similar to phosphorylation, GlcNAcylation (the addition of O-GlcNAc to Ser(Thr) residues on polypeptides) is an abundant, dynamic, and inducible post-translational modification. GlcNAcylated proteins are crucial in regulating virtually all cellular processes, including signaling, cell cycle, and transcription. Here we show that calcium/calmodulin-dependent kinase IV (CaMKIV) is highly GlcNAcylated in vivo. In addition, we show that upon activation of HEK293 cells, hemagglutinin-tagged CaMKIV GlcNAcylation rapidly decreases, in a manner directly opposing its phosphorylation at Thr-200. Correspondingly, there is an increase in CaMKIV interaction with O-GlcNAcase during CaMKIV activation. Furthermore, we identify at least five sites of GlcNAcylation on CaMKIV. Using site-directed mutagenesis, we determine that the GlcNAcylation sites located in the active site of CaMKIV can modulate its phosphorylation at Thr-200 and its activity toward cAMP-response element-binding transcription factor. Our results strongly indicate that the O-GlcNAc modification participates in the regulation of CaMKIV activation and function, possibly coordinating nutritional signals with the immune and nervous systems. This is the first example of an O-GlcNAc/phosphate cycle involving O-GlcNAc transferase/kinase cross-talk.
Collapse
Affiliation(s)
- Wagner B. Dias
- From the Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Win D. Cheung
- From the Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Zihao Wang
- From the Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Gerald W. Hart
- From the Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
32
|
Li H, Liu G, Yu J, Cao W, Lobo VG, Xie J. In vivo selection of kinase-responsive RNA elements controlling alternative splicing. J Biol Chem 2009; 284:16191-16201. [PMID: 19386606 PMCID: PMC2713567 DOI: 10.1074/jbc.m900393200] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 03/23/2009] [Indexed: 12/29/2022] Open
Abstract
Alternative pre-mRNA splicing is often controlled by cell signals, for example, those activating the cAMP-dependent protein kinase (PKA) or the Ca2+/calmodulin-dependent protein kinase IV (CaMKIV). We have shown that CaMKIV regulates alternative splicing through short CA repeats and hnRNP L. Here we use a splicing reporter that shows PKA/CaMKIV promotion of exon inclusion to select from exons containing random 13-nt sequences for RNA elements responsive to the kinases in cultured cells. This selection not only identified both PKA- and CaMKIV-responsive elements that are similar to the CaMKIV-responsive RNA element 1 (CaRRE1) or CA repeats, but also A-rich elements not previously known to respond to these kinases. Consistently, hnRNP L is identified as a factor binding the CA-rich elements. Analyses of the motifs in the highly responsive elements indicate that they are indeed critical for the kinase effect and are enriched in alternative exons. Interestingly, a CAAAAAA motif is sufficient for the PKA/CaMKIV-regulated splicing of the exon 16 of the CaMK kinase beta1 (CaMKK2) transcripts, implying a role of this motif in signaling cross-talk or feedback regulation between these kinases through alternative splicing. Therefore, these experiments identified a group of RNA elements responsive to PKA and CaMKIV from in vivo selection. This also provides an approach for selecting RNA elements similarly responsive to other cell signals controlling alternative splicing.
Collapse
Affiliation(s)
- Hongzhao Li
- From the Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Guodong Liu
- From the Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Jiankun Yu
- From the Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Wenguang Cao
- From the Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Vincent G Lobo
- From the Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Jiuyong Xie
- From the Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.
| |
Collapse
|
33
|
Wang H, Fukushima H, Kida S, Zhuo M. Ca2+/calmodulin-dependent protein kinase IV links group I metabotropic glutamate receptors to fragile X mental retardation protein in cingulate cortex. J Biol Chem 2009; 284:18953-62. [PMID: 19436069 DOI: 10.1074/jbc.m109.019141] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Fragile X syndrome is caused by a lack of fragile X mental retardation protein (FMRP) due to silencing of the FMR1 gene. The metabotropic glutamate receptors (mGluRs) in the central nervous system contribute to higher brain functions including learning/memory, persistent pain, and mental disorders. Our recent study has shown that activation of Group I mGluR up-regulated FMRP in anterior cingulate cortex (ACC), a key region for brain cognitive and executive functions; Ca(2+) signaling pathways could be involved in the regulation of FMRP by Group I mGluRs. In this study we demonstrate that stimulating Group I mGluRs activates Ca(2+)/calmodulin-dependent protein kinase IV (CaMKIV) in ACC neurons. In ACC neurons of adult mice overexpressing CaMKIV, the up-regulation of FMRP by stimulating Group I mGluR is enhanced. The enhancement occurs at the transcriptional level as the Fmr1 mRNA level was further elevated compared with wild-type mice. Using pharmacological approaches, we found that inhibition of CaMKIV could attenuate the up-regulation of FMRP by Group I mGluRs. CaMKIV contribute to the regulation of FMRP by Group I mGluRs probably through cyclic AMP-responsive element binding protein (CREB) activation, as manipulation of CaMKIV could simultaneously cause the change of CREB phosphorylation induced by Group I mGluR activation. Our study has provided strong evidence for CaMKIV as a molecular link between Group I mGluRs and FMRP in ACC neurons and may help us to elucidate the pathogenesis of fragile X syndrome.
Collapse
Affiliation(s)
- Hansen Wang
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | | | | | |
Collapse
|
34
|
Yu J, Hai Y, Liu G, Fang T, Kung SKP, Xie J. The heterogeneous nuclear ribonucleoprotein L is an essential component in the Ca2+/calmodulin-dependent protein kinase IV-regulated alternative splicing through cytidine-adenosine repeats. J Biol Chem 2009; 284:1505-13. [PMID: 19017650 PMCID: PMC3471988 DOI: 10.1074/jbc.m805113200] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of gene expression through alternative pre-mRNA splicing is common in metazoans and is often controlled by intracellular signaling pathways that are important in cell physiology. We have shown that the alternative splicing of a number of genes is controlled by membrane depolarization and Ca2+/calmodulin-dependent protein kinase IV (CaMKIV) through CaMKIV-responsive RNA elements (CaRRE1 and CaRRE2); however, the trans-acting factors remain unknown. Here we show that the heterogeneous nuclear ribonucleoprotein (hnRNP) L is a CaRRE1 binding factor in nuclear extracts. An hnRNP L high affinity CA (cytidine-adenosine) repeat element is sufficient to mediate CaMKIV and hnRNP L repression of splicing in a location (3'-splice site proximity)-dependent way. Depletion of hnRNP L by RNA interference followed by rescue with coexpressed exogenous hnRNP L demonstrates that hnRNP L mediates the CaMKIV-regulated splicing through CA repeats in heterologous contexts. Depletion of hnRNP L also led to increased inclusion of the stress axis-regulated exon and a CA repeat-harboring exon under depolarization or with activated CaMKIV. Moreover, hnRNP L binding to CaRRE1 was increased by CaMKIV and, conversely, was reduced by pretreatments with protein phosphatases. Therefore, hnRNP L is an essential component of CaMKIV-regulated alternative splicing through CA repeats, with its phosphorylation likely playing a critical role.
Collapse
Affiliation(s)
- Jiankun Yu
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Yan Hai
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Guodong Liu
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Tielan Fang
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Sam K. P. Kung
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jiuyong Xie
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
35
|
Zhang X, Wheeler D, Tang Y, Guo L, Shapiro RA, Ribar TJ, Means AR, Billiar TR, Angus DC, Rosengart MR. Calcium/calmodulin-dependent protein kinase (CaMK) IV mediates nucleocytoplasmic shuttling and release of HMGB1 during lipopolysaccharide stimulation of macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:5015-23. [PMID: 18802105 PMCID: PMC2587501 DOI: 10.4049/jimmunol.181.7.5015] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The chromatin-binding factor high-mobility group box 1 (HMGB1) functions as a proinflammatory cytokine and late mediator of mortality in murine endotoxemia. Although serine phosphorylation of HMGB1 is necessary for nucleocytoplasmic shuttling before its cellular release, the protein kinases involved have not been identified. To investigate if calcium/calmodulin-dependent protein kinase (CaMK) IV serine phosphorylates and mediates the release of HMGB1 from macrophages (Mphi) stimulated with LPS, RAW 264.7 cells or murine primary peritoneal Mphi were incubated with either STO609 (a CaMKIV kinase inhibitor), KN93 (a CaMKIV inhibitor), or we utilized cells from which CaMKIV was depleted by RNA interference (RNAi) before stimulation with LPS. We also compared the LPS response of primary Mphi isolated from CaMKIV(+/+) and CaMKIV(-/-) mice. In both cell types LPS induced activation and nuclear translocation of CaMKIV, which preceded HMGB1 nucleocytoplasmic shuttling. However, Mphi treated with KN93, STO609, or CaMKIV RNAi before LPS showed reduced nucleocytoplasmic shuttling of HMGB1 and release of HMGB1 into the supernatant. Additionally, LPS induced serine phosphorylation of HMGB1, which correlated with an interaction between CaMKIV and HMGB1 and with CaMKIV phosphorylation of HMGB1 in vitro. In cells, both HMGB1 phosphorylation and interaction with CaMKIV were inhibited by STO609 or CaMKIV RNAi. Similarly, whereas CaMKIV(+/+) Mphi showed serine phosphorylation of HMGB1 in response to LPS, this phosphorylation was attenuated in CaMKIV(-/-) Mphi. Collectively, our results demonstrate that CaMKIV promotes the nucleocytoplasmic shuttling of HMGB1 and suggest that the process may be mediated through CaMKIV-dependent serine phosphorylation of HMGB1.
Collapse
Affiliation(s)
- Xianghong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - David Wheeler
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Ying Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Lanping Guo
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | | | - Thomas J. Ribar
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Anthony R. Means
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | | | - Derek C. Angus
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Matthew R. Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
36
|
Wayman GA, Lee YS, Tokumitsu H, Silva A, Soderling TR. Calmodulin-kinases: modulators of neuronal development and plasticity. Neuron 2008; 59:914-31. [PMID: 18817731 PMCID: PMC2664743 DOI: 10.1016/j.neuron.2008.08.021] [Citation(s) in RCA: 462] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 08/18/2008] [Accepted: 08/19/2008] [Indexed: 11/26/2022]
Abstract
In the nervous system, many intracellular responses to elevated calcium are mediated by CaM kinases (CaMKs), a family of protein kinases whose activities are initially modulated by binding Ca(2+)/calmodulin and subsequently by protein phosphorylation. One member of this family, CaMKII, is well-established for its effects on modulating synaptic plasticity and learning and memory. However, recent studies indicate that some actions on neuronal development and function attributed to CaMKII may instead or in addition be mediated by other members of the CaMK cascade, such as CaMKK, CaMKI, and CaMKIV. This review summarizes key neuronal functions of the CaMK cascade in signal transduction, gene transcription, synaptic development and plasticity, and behavior. The technical challenges of mapping cellular protein kinase signaling pathways are also discussed.
Collapse
Affiliation(s)
- Gary A. Wayman
- Vollum Institute, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Yong-Seok Lee
- Department of Neurobiology, University of California, Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Hiroshi Tokumitsu
- Department of Signal Transduction Sciences, Kagawa University, Kagawa 761-0793, Japan
| | - Alcino Silva
- Department of Neurobiology, University of California, Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Thomas R. Soderling
- Vollum Institute, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
37
|
Braconi Quintaje S, Orchard S. The annotation of both human and mouse kinomes in UniProtKB/Swiss-Prot: one small step in manual annotation, one giant leap for full comprehension of genomes. Mol Cell Proteomics 2008; 7:1409-19. [PMID: 18436524 PMCID: PMC2500232 DOI: 10.1074/mcp.r700001-mcp200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 04/17/2008] [Indexed: 01/09/2023] Open
Abstract
Biomolecule phosphorylation by protein kinases is a fundamental cell signaling process in all living cells. Following the comprehensive cataloguing of the protein kinase complement of the human genome (Manning, G., Whyte, D. B., Martinez, R., Hunter, T., and Sudarsanam, S. (2002) The protein kinase complement of the human genome. Science 298, 1912-1934), this review will detail the state-of-the-art human and mouse kinase proteomes as provided in the UniProtKB/Swiss-Prot protein knowledgebase. The sequences of the 480 classical and up to 24 atypical protein kinases now believed to exist in the human genome and 484 classical and up to 24 atypical kinases within the mouse genome have been reviewed and, where necessary, revised. Extensive annotation has been added to each entry. In an era when a wealth of new databases is emerging on the Internet, UniProtKB/Swiss-Prot makes available to the scientific community the most up-to-date and in-depth annotation of these proteins with access to additional external resources linked from within each entry. Incorrect sequence annotations resulting from errors and artifacts have been eliminated. Each entry will be constantly reviewed and updated as new information becomes available with the orthologous enzymes in related species being annotated in a parallel effort and complete kinomes being completed as sequences become available. This ensures that the mammalian kinomes available from UniProtKB/Swiss-Prot are of a consistently high standard with each separate entry acting both as a valuable information resource and a central portal to a wealth of further detail via extensive cross-referencing.
Collapse
Affiliation(s)
- Silvia Braconi Quintaje
- Swiss-Prot group, Swiss Institute of Bioinformatics, CMU, 1 Michel Servet, 1211 Geneva 4, Switzerland
| | | |
Collapse
|
38
|
Gaines P, Lamoureux J, Marisetty A, Chi J, Berliner N. A cascade of Ca(2+)/calmodulin-dependent protein kinases regulates the differentiation and functional activation of murine neutrophils. Exp Hematol 2008; 36:832-44. [PMID: 18400360 PMCID: PMC2577899 DOI: 10.1016/j.exphem.2008.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 01/28/2008] [Accepted: 02/14/2008] [Indexed: 01/21/2023]
Abstract
OBJECTIVE The function of neutrophils as primary mediators of innate immunity depends on the activity of granule proteins and critical components of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. Expression of their cognate genes is regulated during neutrophil differentiation by a complex network of intracellular signaling pathways. In this study, we have investigated the role of two members of the calcium/calmodulin-dependent protein kinase (CaMK) signaling cascade, CaMK I-like kinase (CKLiK) and CaMKKalpha, in regulating neutrophil differentiation and functional activation. MATERIALS AND METHODS Mouse myeloid cell lines were used to examine the expression of a CaMK cascade in developing neutrophils and to examine the effects of constitutive activation vs inhibition of CaMKs on neutrophil maturation. RESULTS Expression of CaMKKalpha was shown to increase during neutrophil differentiation in multiple cell lines, whereas expression of CKLiK increased as multipotent progenitors committed to promyelocytes, but then decreased as cells differentiated into mature neutrophils. Expression of constitutively active CKLiKs did not affect morphologic maturation, but caused dramatic decreases in both respiratory burst responses and chemotaxis. This loss of neutrophil function was accompanied by reduced secondary granule and gp91(phox) gene expression. The CaMK inhibitor KN-93 attenuated cytokine-stimulated proliferative responses in promyelocytic cell lines, and inhibited the respiratory burst. Similar data were observed with the CaMKKalpha inhibitor, STO-609. CONCLUSIONS Overactivation of a cascade of CaMKs inhibits neutrophil maturation, suggesting that these kinases play an antagonistic role during neutrophil differentiation, but at least one CaMK is required for myeloid cell expansion and functional activation.
Collapse
Affiliation(s)
- Peter Gaines
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA.
| | | | | | | | | |
Collapse
|
39
|
Spencer TK, Mellado W, Filbin MT. BDNF activates CaMKIV and PKA in parallel to block MAG-mediated inhibition of neurite outgrowth. Mol Cell Neurosci 2008; 38:110-6. [PMID: 18381242 PMCID: PMC2692908 DOI: 10.1016/j.mcn.2008.02.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 02/13/2008] [Indexed: 12/12/2022] Open
Abstract
The environment of the adult CNS prevents axonal regeneration after injury. This inhibition of axonal regeneration can be blocked by elevating cAMP. Previously, we showed that the cAMP pathway can be activated via pre-treatment with neurotrophins and requires activation of several signaling pathways which converge at activation of the transcription factor, CREB. Here, we show that calcium/calmodulin-dependent kinase IV (CaMKIV) is necessary for the neurotrophin-induced phosphorylation of CREB and the block of myelin-mediated inhibition of axonal growth. Pharmacological inhibition of CaMKIV or over-expression of a dominant-negative mutant form of CaMKIV blocks the neurotrophin effect. Interestingly, CaMKIV activation is not necessary if cAMP levels is already elevated. Finally, calcium flux from intracellular stores is necessary for this CaMKIV signaling. These results demonstrate that CaMKIV is another player in the neurotrophin-induced signaling which leads to axonal regeneration and therefore, is a potential target for therapeutic intervention following injury to the adult CNS.
Collapse
Affiliation(s)
- Timothy K Spencer
- Department of Biological Sciences, Hunter College, City University of New York, New York, New York 10065, USA.
| | | | | |
Collapse
|
40
|
Ibata K, Sun Q, Turrigiano GG. Rapid synaptic scaling induced by changes in postsynaptic firing. Neuron 2008; 57:819-26. [PMID: 18367083 DOI: 10.1016/j.neuron.2008.02.031] [Citation(s) in RCA: 344] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 04/27/2007] [Accepted: 02/29/2008] [Indexed: 12/17/2022]
Abstract
Homeostatic synaptic scaling adjusts a neuron's excitatory synaptic strengths up or down to compensate for perturbations in activity. Little is known about the molecular pathway(s) involved, nor is it clear which aspect of "activity"-local synaptic signaling, postsynaptic firing, or large-scale changes in network activity-is required to induce synaptic scaling. Here, we selectively block either postsynaptic firing in individual neurons or a fraction of presynaptic inputs, while optically monitoring changes in synaptic strength. We find that synaptic scaling is rapidly induced by block of postsynaptic firing, but not by local synaptic blockade, and is mediated through a drop in somatic calcium influx, reduced activation of CaMKIV, and an increase in transcription. Cortical neurons thus homeostatically adjust synaptic strengths in response to changes in their own firing rate, a mechanism with the computational advantage of efficiently normalizing synaptic strengths without interfering with synapse-specific mechanisms of information storage.
Collapse
Affiliation(s)
- Keiji Ibata
- Department of Biology and Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | | | | |
Collapse
|
41
|
Colomer J, Means AR. Physiological roles of the Ca2+/CaM-dependent protein kinase cascade in health and disease. Subcell Biochem 2008; 45:169-214. [PMID: 18193638 DOI: 10.1007/978-1-4020-6191-2_7] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Numerous hormones, growth factors and physiological processes cause a rise in cytosolic Ca2+, which is translated into meaningful cellular responses by interacting with a large number of Ca2(+)-binding proteins. The Ca2(+)-binding protein that is most pervasive in mediating these responses is calmodulin (CaM), which acts as a primary receptor for Ca2+ in all eukaryotic cells. In turn, Ca2+/CaM functions as an allosteric activator of a host of enzymatic proteins including a considerable number of protein kinases. The topic of this review is to discuss the physiological roles of a sub-set of these protein kinases which can function in cells as a Ca2+/CaM-dependent kinase signaling cascade. The cascade was originally believed to consist of a CaM kinase kinase that phosphorylates and activates one of two CaM kinases, CaMKI or CaMKIV. The unusual aspect of this cascade is that both the kinase kinase and the kinase require the binding of Ca2+/CaM for activation. More recently, one of the CaM kinase kinases has been found to activate another important enzyme, the AMP-dependent protein kinase so the concept of the CaM kinase cascade must be expanded. A CaM kinase cascade is important for many normal physiological processes that when misregulated can lead to a variety of disease states. These processes include: cell proliferation and apoptosis that may conspire in the genesis of cancer; neuronal growth and function related to brain development, synaptic plasticity as well as memory formation and maintenance; proper function of the immune system including the inflammatory response, activation of T lymphocytes and hematopoietic stem cell maintenance; and the central control of energy balance that, when altered, can lead to obesity and diabetes. Although the study of the CaM-dependent kinase cascades is still in its infancy continued analysis of the pathways regulated by these Ca2(+)-initiated signaling cascades holds considerable promise for the future of disease-related research.
Collapse
Affiliation(s)
- J Colomer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center USA
| | | |
Collapse
|
42
|
Smith JAH, Collins M, Grobler LA, Magee CJ, Ojuka EO. Exercise and CaMK activation both increase the binding of MEF2A to the Glut4 promoter in skeletal muscle in vivo. Am J Physiol Endocrinol Metab 2007; 292:E413-20. [PMID: 16985263 DOI: 10.1152/ajpendo.00142.2006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In vitro binding assays have indicated that the exercise-induced increase in muscle GLUT4 is preceded by increased binding of myocyte enhancer factor 2A (MEF2A) to its cis-element on the Glut4 promoter. Because in vivo binding conditions are often not adequately recreated in vitro, we measured the amount of MEF2A that was bound to the Glut4 promoter in rat triceps after an acute swimming exercise in vivo, using chromatin immunoprecipitation (ChIP) assays. Bound MEF2A was undetectable in nonexercised controls or at 24 h postexercise but was significantly elevated approximately 6 h postexercise. Interestingly, the increase in bound MEF2A was preceded by an increase in autonomous activity of calcium/calmodulin-dependent protein kinase (CaMK) II in the same muscle. To determine if CaMK signaling mediates MEF2A/DNA associations in vivo, we performed ChIP assays on C(2)C(12) myotubes expressing constitutively active (CA) or dominant negative (DN) CaMK IV proteins. We found that approximately 75% more MEF2A was bound to the Glut4 promoter in CA compared with DN CaMK IV-expressing cells. GLUT4 protein increased approximately 70% 24 h after exercise but was unchanged by overexpression of CA CaMK IV in myotubes. These results confirm that exercise increases the binding of MEF2A to the Glut4 promoter in vivo and provides evidence that CaMK signaling is involved in this interaction.
Collapse
Affiliation(s)
- James A H Smith
- UCT/MRC Research Unit for Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, P.O. Box 115, Newlands 7725, South Africa
| | | | | | | | | |
Collapse
|
43
|
Taylor DM, De Koninck P, Minotti S, Durham HD. Manipulation of protein kinases reveals different mechanisms for upregulation of heat shock proteins in motor neurons and non-neuronal cells. Mol Cell Neurosci 2007; 34:20-33. [PMID: 17113785 DOI: 10.1016/j.mcn.2006.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 09/12/2006] [Accepted: 09/27/2006] [Indexed: 01/13/2023] Open
Abstract
Motor neurons have a high threshold for induction of heat shock proteins (Hsps) in response to stress, a property associated with impaired ability to activate heat shock transcription factor 1 (Hsf1). Hyperphosphorylation of Hsf1 has been established as a requirement for transactivation of heat shock genes. This study demonstrated that the impaired heat shock response in motor neurons is not due to altered phosphorylation of Hsf1 by kinases previously shown to affect activation of Hsf1 in other cells (PKC, GSK3beta, ERK1, CaMKIIalpha). However, a constitutively active form of CaMKIV induced robust expression of Hsp70, as well as transcription of a GFP reporter gene driven by the human inducible Hsp70 promoter in unstressed motor neurons, but not in mouse embryonic fibroblasts. The results point to novel mechanisms of activation of heat shock genes in motor neurons that have relevance to exploitation of endogenous stress responses therapeutically.
Collapse
Affiliation(s)
- David M Taylor
- Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, Canada H3A 2B4
| | | | | | | |
Collapse
|
44
|
The calcium/calmodulin-dependent protein kinase cascades. CALCIUM - A MATTER OF LIFE OR DEATH 2007. [DOI: 10.1016/s0167-7306(06)41013-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Blaeser F, Sanders MJ, Truong N, Ko S, Wu LJ, Wozniak DF, Fanselow MS, Zhuo M, Chatila TA. Long-term memory deficits in Pavlovian fear conditioning in Ca2+/calmodulin kinase kinase alpha-deficient mice. Mol Cell Biol 2006; 26:9105-15. [PMID: 17015467 PMCID: PMC1636841 DOI: 10.1128/mcb.01452-06] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Signaling by the Ca(2+)/calmodulin kinase (CaMK) cascade has been implicated in neuronal gene transcription, synaptic plasticity, and long-term memory consolidation. The CaM kinase kinase alpha (CaMKKalpha) isoform is an upstream component of the CaMK cascade whose function in different behavioral and learning and memory paradigms was analyzed by targeted gene disruption in mice. CaMKKalpha mutants exhibited normal long-term spatial memory formation and cued fear conditioning but showed deficits in context fear during both conditioning and long-term follow-up testing. They also exhibited impaired activation of the downstream kinase CaMKIV/Gr and its substrate, the transcription factor cyclic AMP-responsive element binding protein (CREB) upon fear conditioning. Unlike CaMKIV/Gr-deficient mice, the CaMKKalpha mutants exhibited normal long-term potentiation and normal levels of anxiety-like behavior. These results demonstrate a selective role for CaMKKalpha in contextual fear memory and suggest that different combinations of upstream and downstream components of the CaMK cascade may serve distinct physiological functions.
Collapse
Affiliation(s)
- Frank Blaeser
- Department of Pediatrics, The David Geffen School of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095-1752, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mercy L, Pauw AD, Payen L, Tejerina S, Houbion A, Demazy C, Raes M, Renard P, Arnould T. Mitochondrial biogenesis in mtDNA-depleted cells involves a Ca2+-dependent pathway and a reduced mitochondrial protein import. FEBS J 2005; 272:5031-55. [PMID: 16176275 DOI: 10.1111/j.1742-4658.2005.04913.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Alterations in mitochondrial activity resulting from defects in mitochondrial DNA (mtDNA) can modulate the biogenesis of mitochondria by mechanisms that are still poorly understood. In order to study mitochondrial biogenesis in cells with impaired mitochondrial activity, we used rho-L929 and rho(0)143 B cells (partially and totally depleted of mtDNA, respectively), that maintain and even up-regulate mitochondrial population, to characterize the activity of major transcriptional regulators (Sp1, YY1, MEF2, PPARgamma, NRF-1, NRF-2, CREB and PGC-1alpha) known to control the expression of numerous nuclear genes encoding mitochondrial proteins. Among these regulators, cyclic AMP-responsive element binding protein (CREB) activity was the only one to be increased in mtDNA-depleted cells. CREB activation mediated by a calcium-dependent pathway in these cells also regulates the expression of cytochrome c and the abundance of mitochondrial population as both are decreased in mtDNA-depleted cells that over-express CREB dominant negative mutants. Mitochondrial biogenesis in mtDNA-depleted cells is also dependent on intracellular calcium as its chelation reduces mitochondrial mass. Despite a slight increase in mitochondrial mass in mtDNA-depleted cells, the mitochondrial protein import activity was reduced as shown by a decrease in the import of radiolabeled matrix-targeted recombinant proteins into isolated mitochondria and by the reduced mitochondrial localization of ectopically expressed HA-apoaequorin targeted to the mitochondria. Decrease in ATP content, in mitochondrial membrane potential as well as reduction in mitochondrial Tim44 abundance could explain the lower mitochondrial protein import in mtDNA-depleted cells. Taken together, these results suggest that mitochondrial biogenesis is stimulated in mtDNA-depleted cells and involves a calcium-CREB signalling pathway but is associated with a reduced mitochondrial import for matrix proteins.
Collapse
Affiliation(s)
- Ludovic Mercy
- Laboratory of Biochemistry and Cellular Biology, University of Namur (FUNDP), Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ellison TI, Dowd DR, MacDonald PN. Calmodulin-Dependent Kinase IV Stimulates Vitamin D Receptor-Mediated Transcription. Mol Endocrinol 2005; 19:2309-19. [PMID: 15919723 DOI: 10.1210/me.2004-0382] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] promotes intestinal absorption of calcium primarily by binding to the vitamin D receptor (VDR) and regulating gene expression. 1,25-(OH)2D3 also exerts rapid actions at the cell membrane that include increasing intracellular calcium levels and activating protein kinase cascades. To explore potential cross talk between calcium signaling elicited by the nongenomic actions of 1,25-(OH)2D3 and the genomic pathway mediated by VDR, we examined the effects of activated Ca2+/calmodulin-dependent kinases (CaMKs) on 1,25-(OH)2D3/VDR-mediated transcription. Expression of a constitutively active form of CaMKIV dramatically stimulated 1,25-(OH)2D3-activated reporter gene expression in COS-7, HeLa, and ROS17/2.8 cell lines. Metabolic labeling studies indicated that CaMKIV increased VDR phosphorylation levels. In addition, CaMKIV increased the independent transcription activity of the VDR coactivator SRC (steroid receptor coactivator) 1, and promoted ligand-dependent interaction between VDR and SRC coactivator proteins in mammalian two-hybrid studies. The functional consequences of this multifaceted mechanism of CaMKIV action were revealed by reporter gene studies, which showed that CaMKIV and select SRC coactivators synergistically enhanced VDR-mediated transcription. These studies support a model in which CaMKIV signaling stimulates VDR-mediated transcription by increasing phosphorylation levels of VDR and enhancing autonomous SRC activity, resulting in higher 1,25-(OH)2D3-dependent interaction between VDR and SRC coactivators.
Collapse
Affiliation(s)
- Tara I Ellison
- Department of Pharmacology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | | | | |
Collapse
|
48
|
Juang YT, Wang Y, Solomou EE, Li Y, Mawrin C, Tenbrock K, Kyttaris VC, Tsokos GC. Systemic lupus erythematosus serum IgG increases CREM binding to the IL-2 promoter and suppresses IL-2 production through CaMKIV. J Clin Invest 2005. [PMID: 15841182 DOI: 10.1172/jci200522854] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) T cells express high levels of cAMP response element modulator (CREM) that binds to the IL-2 promoter and represses the transcription of the IL-2 gene. This study was designed to identify pathways that lead to increased binding of CREM to the IL-2 promoter in SLE T cells. Ca(2+)/calmodulin-dependent kinase IV (CaMKIV) was found to be increased in the nucleus of SLE T cells and to be involved in the overexpression of CREM and its binding to the IL-2 promoter. Treatment of normal T cells with SLE serum resulted in increased expression of CREM protein, increased binding of CREM to the IL-2 promoter, and decreased IL-2 promoter activity and IL-2 production. This process was abolished when a dominant inactive form of CaMKIV was expressed in normal T cells. The effect of SLE serum resided within the IgG fraction and was specifically attributed to anti-TCR/CD3 autoantibodies. This study identifies CaMKIV as being responsible for the increased expression of CREM and the decreased production of IL-2 in SLE T cells and demonstrates that anti-TCR/CD3 antibodies present in SLE sera can account for the increased expression of CREM and the suppression of IL-2 production.
Collapse
Affiliation(s)
- Yuang-Taung Juang
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Pan F, Means AR, Liu JO. Calmodulin-dependent protein kinase IV regulates nuclear export of Cabin1 during T-cell activation. EMBO J 2005; 24:2104-13. [PMID: 15902271 PMCID: PMC1150881 DOI: 10.1038/sj.emboj.7600685] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Accepted: 04/27/2005] [Indexed: 11/08/2022] Open
Abstract
Calcium signaling is critical for activation of T lymphocytes and has been proposed to be transduced through multiple calmodulin target proteins. Whereas the calcineurin-NFAT signaling module is critical for all mammalian T cells, the role of calmodulin-dependent kinase IV (CaMKIV) in mouse naïve CD4+ T-cell activation remains enigmatic. We have applied lentivius-mediated RNA interference of CaMKIV to human T cells and found that knockdown of CaMKIV abrogates T-cell receptor-mediated transcription of the IL-2 gene. We demonstrate that CaMKIV directly phosphorylates Cabin1, a transcriptional corepressor for myocyte enhancer factor 2, creating a docking site for 14-3-3, which causes its nuclear export. CaMKIV-mediated nuclear export of Cabin1 is likely to account for a significant part of the requirement of CaMKIV during human T-cell activation.
Collapse
Affiliation(s)
- Fan Pan
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony R Means
- Departments of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Jun O Liu
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Johns Hopkins University School of Medicine, 725 North Wolf Street, Baltimore, MD 21205, USA. Tel.: +1 410 955 4619; Fax: +1 410 955 4620; E-mail:
| |
Collapse
|
50
|
Verploegen S, Ulfman L, van Deutekom HWM, van Aalst C, Honing H, Lammers JWJ, Koenderman L, Coffer PJ. Characterization of the role of CaMKI-like kinase (CKLiK) in human granulocyte function. Blood 2005; 106:1076-83. [PMID: 15840691 DOI: 10.1182/blood-2004-09-3755] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of granulocyte effector functions, such as induction of the respiratory burst and migration, are regulated by a variety of relatively ill-defined signaling pathways. Recently, we identified a novel Ca2+/calmodulin-dependent kinase I-like kinase, CKLiK, which exhibits restricted mRNA expression to human granulocytes. Using a novel antibody generated against the C-terminus of CKLiK, CKLiK was detected in CD34+-derived neutrophils and eosinophils, as well as in mature peripheral blood granulocytes. Activation of human granulocytes by N-formyl-methionyl-leucyl-phenylalanine (fMLP) and platelet-activating factor (PAF), but not the phorbol ester PMA (phorbol 12-myristate-13-acetate), resulted in induction of CKLiK activity, in parallel with a rise of intracellular Ca2+ [Ca2+]i. To study the functionality of CKLiK in human granulocytes, a cell-permeable CKLiK peptide inhibitor (CKLiK297-321) was generated which was able to inhibit kinase activity in a dose-dependent manner. The effect of this peptide was studied on specific granulocyte effector functions such as phagocytosis, respiratory burst, migration, and adhesion. Phagocytosis of Aspergillus fumigatus particles was reduced in the presence of CKLiK297-321 and fMLP-induced reactive oxygen species (ROS) production was potently inhibited by CKLiK297-321 in a dose-dependent manner. Furthermore, fMLP-induced neutrophil migration on albumin-coated surfaces was perturbed, as well as beta2-integrin-mediated adhesion. These findings suggest a critical role for CKLiK in modulating chemoattractant-induced functional responses in human granulocytes.
Collapse
Affiliation(s)
- Sandra Verploegen
- Molecular Immunology Lab, Department of Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|