1
|
Chen Y, Luo M, Tu H, Qi Y, Guo Y, Zhang X, Cui Y, Gao M, Zhou X, Zhu T, Zhu H, Situ C, Li Y, Guo X. STYXL1 regulates CCT complex assembly and flagellar tubulin folding in sperm formation. Nat Commun 2024; 15:44. [PMID: 38168070 PMCID: PMC10761714 DOI: 10.1038/s41467-023-44337-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Tubulin-based microtubule is a core component of flagella axoneme and essential for sperm motility and male fertility. Structural components of the axoneme have been well explored. However, how tubulin folding is regulated in sperm flagella formation is still largely unknown. Here, we report a germ cell-specific co-factor of CCT complex, STYXL1. Deletion of Styxl1 results in male infertility and microtubule defects of sperm flagella. Proteomic analysis of Styxl1-/- sperm reveals abnormal downregulation of flagella-related proteins including tubulins. The N-terminal rhodanese-like domain of STYXL1 is important for its interactions with CCT complex subunits, CCT1, CCT6 and CCT7. Styxl1 deletion leads to defects in CCT complex assembly and tubulin polymerization. Collectively, our findings reveal the vital roles of germ cell-specific STYXL1 in CCT-facilitated tubulin folding and sperm flagella development.
Collapse
Affiliation(s)
- Yu Chen
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
- Medical Research Center, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China
| | - Mengjiao Luo
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Haixia Tu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Yaling Qi
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yueshuai Guo
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiangzheng Zhang
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yiqiang Cui
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mengmeng Gao
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xin Zhou
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Tianyu Zhu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hui Zhu
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chenghao Situ
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yan Li
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China.
| | - Xuejiang Guo
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
2
|
Tang H, Peng T, Yang X, Liu L, Xu Y, Zhao Y, Huang S, Fu C, Huang Y, Zhou H, Li J, He L, Wang W, Niu H, Xu K. Plasma Metabolomic Changes in Children with Cerebral Palsy Exposed to Botulinum Neurotoxin. J Proteome Res 2022; 21:671-682. [PMID: 35018779 DOI: 10.1021/acs.jproteome.1c00711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The long-term effect of botulinum neurotoxin A (BoNT-A) on children with cerebral palsy (CP) is unclear, and how the dynamic changes of metabolites impact the duration of effect remains unknown. To tackle this, we collected 120 plasma samples from 91 children with spastic CP for analysis, with 30 samples in each time point: prior to injection and 1, 3, and 6 months after injection. A total of 354 metabolites were identified across all the time points, 39 of which exhibited significant changes (with tentative IDs) (p values <0.05, VIP > 1). Principal component analysis and partial least-squares discriminant analysis disclosed a clear separation between different groups (p values <0.05). Network analysis revealed the coordinated changes of functional metabolites. Pathway analysis highlighted the metabolic pathways associated with energy consumption and glycine, serine, and threonine metabolism and cysteine and methionine metabolism. Collectively, our results identified the significant dynamic changes of plasma metabolite after BoNT-A injections on children with CP. Metabolic pathways associated with energy expenditure might provide a new perspective for the effect of BoNT-A in children with CP. Glycine, serine, and threonine metabolism and cysteine and methionine metabolism might be related to the duration of effect of BoNT-A.
Collapse
Affiliation(s)
- Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Yunxian Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Yiting Zhao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Shiya Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Chaoqiong Fu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Yuan Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China.,Department of Rehabilitation, School of Medicine, South China University of Technology, Guangzhou 510655, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Jinling Li
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Wenda Wang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| | - Huiran Niu
- Genechem Biotechnology Co., Ltd. Shanghai 200120, China
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, Guangzhou China
| |
Collapse
|
3
|
Cilleros-Mañé V, Just-Borràs L, Polishchuk A, Durán M, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. M 1 and M 2 mAChRs activate PDK1 and regulate PKC βI and ε and the exocytotic apparatus at the NMJ. FASEB J 2021; 35:e21724. [PMID: 34133802 DOI: 10.1096/fj.202002213r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Neuromuscular junctions (NMJ) regulate cholinergic exocytosis through the M1 and M2 muscarinic acetylcholine autoreceptors (mAChR), involving the crosstalk between receptors and downstream pathways. Protein kinase C (PKC) regulates neurotransmission but how it associates with the mAChRs remains unknown. Here, we investigate whether mAChRs recruit the classical PKCβI and the novel PKCε isoforms and modulate their priming by PDK1, translocation and activity on neurosecretion targets. We show that each M1 and M2 mAChR activates the master kinase PDK1 and promotes a particular priming of the presynaptic PKCβI and ε isoforms. M1 recruits both primed-PKCs to the membrane and promotes Munc18-1, SNAP-25, and MARCKS phosphorylation. In contrast, M2 downregulates PKCε through a PKA-dependent pathway, which inhibits Munc18-1 synthesis and PKC phosphorylation. In summary, our results discover a co-dependent balance between muscarinic autoreceptors which orchestrates the presynaptic PKC and their action on ACh release SNARE-SM mechanism. Altogether, this molecular signaling explains previous functional studies at the NMJ and guide toward potential therapeutic targets.
Collapse
Affiliation(s)
- V Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - L Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - A Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Durán
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - N Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - J M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
4
|
Jin Y, Zhang C, Fang X, Fang C, Chen J, Du R, Hu Q, Dong L, Zhu Z, Wang T. SNAP25 protects primary cortical neurons from hypoxic-ischemic injury associated with CREB signal. IBRAIN 2021; 7:1-11. [PMID: 37786874 PMCID: PMC10528992 DOI: 10.1002/j.2769-2795.2021.tb00058.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 10/04/2023]
Abstract
Background Hypoxic-ischemic encephalopathy (HIE) could induce exacerbated changes and unpredictable effects in brain cells, and the mechanism remains unclear. Methods HIE model was established in neonatal rats, Zea-Longa score and TTC staining were used to observe the neurobehavior and brain infarct volume in rats subjected to cerebral hypoxia-ischemia (HI). Primary cortical neurons were then cultured in vitro to establish an oxygen and glucose deprivation model. To determine the role of synaptosomal-associated protein-25 (SNAP25) in HIE, PC12 cells were cultured and effective siRNA fragments were screened, and SNAP25 was transfected into primary neurons. Then, quantitative real-time polymerase chain reaction was used to detect the mRNA expression level and immunofluorescence staining was used to observe the morphological changes of neurons before and after the injury. Finally, the abundance values of SNAP25 and its associated genes were filtered using the NCBI and GeneMANIA, respectively. Results HI leads to a decrease in neuronal number and an increase in SNAP25 expression. Whereas, the interference of SNAP25 caused marked decrease in neuronal number and the length of neurite. Moreover, the expression levels of CREB and SYP were significantly decreased after interference of SNAP25. Conclusion SNAP25 exhibited several neuroprotective effects to neuronal protection in neonatal cerebral HI by regulating CREB and SYP.
Collapse
Affiliation(s)
- Yuan Jin
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| | - Chao Zhang
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Xu Fang
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Chang‐Le Fang
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research CenterDepartment of AnesthesiologyDepartment of Cardiovascular DiseaseAffiliated Traditional Chinese Medicine Hospital, Southwest Medical UniversityLuzhouSichuanChina
| | - Jie Chen
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research CenterDepartment of AnesthesiologyDepartment of Cardiovascular DiseaseAffiliated Traditional Chinese Medicine Hospital, Southwest Medical UniversityLuzhouSichuanChina
| | - Ruo‐Lan Du
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| | - Qiao Hu
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| | - Liang Dong
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Zhao‐Qiong Zhu
- Department of AnesthesiologyGuizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Ting‐Hua Wang
- Institute of Neuroscience, Kunming Medical UniveristyKunmingYunnanChina
| |
Collapse
|
5
|
The SNAP-25 Protein Family. Neuroscience 2019; 420:50-71. [DOI: 10.1016/j.neuroscience.2018.09.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/31/2018] [Accepted: 09/14/2018] [Indexed: 01/04/2023]
|
6
|
Simó A, Cilleros-Mañé V, Just-Borràs L, Hurtado E, Nadal L, Tomàs M, Garcia N, Lanuza MA, Tomàs J. nPKCε Mediates SNAP-25 Phosphorylation of Ser-187 in Basal Conditions and After Synaptic Activity at the Neuromuscular Junction. Mol Neurobiol 2019; 56:5346-5364. [PMID: 30607888 DOI: 10.1007/s12035-018-1462-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022]
Abstract
Protein kinase C (PKC) and substrates like SNAP-25 regulate neurotransmission. At the neuromuscular junction (NMJ), PKC promotes neurotransmitter release during synaptic activity. Thirty minutes of muscle contraction enhances presynaptic PKC isoform levels, specifically cPKCβI and nPKCε, through retrograde BDNF/TrkB signaling. This establishes a larger pool of these PKC isoforms ready to promote neuromuscular transmission. The PKC phosphorylation site in SNAP-25 has been mapped to the serine 187 (Ser-187), which is known to enhance calcium-dependent neurotransmitter release in vitro. Here, we localize SNAP-25 at the NMJ and investigate whether cPKCβI and/or nPKCε regulate SNAP-25 phosphorylation. We also investigate whether nerve and muscle cell activities regulate differently SNAP-25 phosphorylation and the involvement of BDNF/TrkB signaling. Our results demonstrate that nPKCε isoform is essential to positively regulate SNAP-25 phosphorylation on Ser-187 and that muscle contraction prevents it. TrkB and cPKCβI do not regulate SNAP-25 protein level or its phosphorylation during neuromuscular activity. The results provide evidence that nerve terminals need both pre- and postsynaptic activities to modulate SNAP-25 phosphorylation and ensure an accurate neurotransmission process.
Collapse
Affiliation(s)
- Anna Simó
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Victor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
7
|
Diverse exocytic pathways for mast cell mediators. Biochem Soc Trans 2018; 46:235-247. [PMID: 29472369 DOI: 10.1042/bst20170450] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/23/2017] [Accepted: 01/04/2018] [Indexed: 12/14/2022]
Abstract
Mast cells play pivotal roles in innate and adaptive immunities but are also culprits in allergy, autoimmunity, and cardiovascular diseases. Mast cells respond to environmental changes by initiating regulated exocytosis/secretion of various biologically active compounds called mediators (e.g. proteases, amines, and cytokines). Many of these mediators are stored in granules/lysosomes and rely on intricate degranulation processes for release. Mast cell stabilizers (e.g. sodium cromoglicate), which prevent such degranulation processes, have therefore been clinically employed to treat asthma and allergic rhinitis. However, it has become increasingly clear that different mast cell diseases often involve multiple mediators that rely on overlapping but distinct mechanisms for release. This review illustrates existing evidence that highlights the diverse exocytic pathways in mast cells. We also discuss strategies to delineate these pathways so as to identify unique molecular components which could serve as new drug targets for more effective and specific treatments against mast cell-related diseases.
Collapse
|
8
|
SNAP-25 phosphorylation at Ser187 regulates synaptic facilitation and short-term plasticity in an age-dependent manner. Sci Rep 2017; 7:7996. [PMID: 28801590 PMCID: PMC5554206 DOI: 10.1038/s41598-017-08237-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/10/2017] [Indexed: 11/18/2022] Open
Abstract
Neurotransmitter release is mediated by the SNARE complex, but the role of its phosphorylation has scarcely been elucidated. Although PKC activators are known to facilitate synaptic transmission, there has been a heated debate on whether PKC mediates facilitation of neurotransmitter release through phosphorylation. One of the SNARE proteins, SNAP-25, is phosphorylated at the residue serine-187 by PKC, but its physiological significance has been unclear. To examine these issues, we analyzed mutant mice lacking the phosphorylation of SNAP-25 serine-187 and found that they exhibited reduced release probability and enhanced presynaptic short-term plasticity, suggesting that not only the release process, but also the dynamics of synaptic vesicles was regulated by the phosphorylation. Furthermore, it has been known that the release probability changes with development, but the precise mechanism has been unclear, and we found that developmental changes in release probability of neurotransmitters were regulated by the phosphorylation. These results indicate that SNAP-25 phosphorylation developmentally facilitates neurotransmitter release but strongly inhibits presynaptic short-term plasticity via modification of the dynamics of synaptic vesicles in presynaptic terminals.
Collapse
|
9
|
Ardito F, Giuliani M, Perrone D, Troiano G, Lo Muzio L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (Review). Int J Mol Med 2017; 40:271-280. [PMID: 28656226 PMCID: PMC5500920 DOI: 10.3892/ijmm.2017.3036] [Citation(s) in RCA: 828] [Impact Index Per Article: 103.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022] Open
Abstract
Protein phosphorylation is an important cellular regulatory mechanism as many enzymes and receptors are activated/deactivated by phosphorylation and dephosphorylation events, by means of kinases and phosphatases. In particular, the protein kinases are responsible for cellular transduction signaling and their hyperactivity, malfunction or overexpression can be found in several diseases, mostly tumors. Therefore, it is evident that the use of kinase inhibitors can be valuable for the treatment of cancer. In this review, we discuss the mechanism of action of phosphorylation, with particular attention to the importance of phosphorylation under physiological and pathological conditions. We also discuss the possibility of using kinase inhibitors in the treatment of tumors.
Collapse
Affiliation(s)
- Fatima Ardito
- Department of Clinical and Experimental Medicine, Foggia University, I-71122 Foggia, Italy
| | - Michele Giuliani
- Department of Clinical and Experimental Medicine, Foggia University, I-71122 Foggia, Italy
| | - Donatella Perrone
- Department of Clinical and Experimental Medicine, Foggia University, I-71122 Foggia, Italy
| | - Giuseppe Troiano
- Department of Clinical and Experimental Medicine, Foggia University, I-71122 Foggia, Italy
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, Foggia University, I-71122 Foggia, Italy
| |
Collapse
|
10
|
Wang J, Casals-Diaz L, Zurawski T, Meng J, Moriarty O, Nealon J, Edupuganti OP, Dolly O. A novel therapeutic with two SNAP-25 inactivating proteases shows long-lasting anti-hyperalgesic activity in a rat model of neuropathic pain. Neuropharmacology 2017; 118:223-232. [PMID: 28347837 DOI: 10.1016/j.neuropharm.2017.03.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/16/2017] [Accepted: 03/23/2017] [Indexed: 01/08/2023]
Abstract
A pressing need exists for long-acting, non-addictive medicines to treat chronic pain, a major societal burden. Botulinum neurotoxin type A (BoNT/A) complex - a potent, specific and prolonged inhibitor of neuro-exocytosis - gives some relief in several pain disorders, but not for all patients. Our study objective was to modify BoNT/A to overcome its inability to block transmitter release elicited by high [Ca2+]i and increase its limited analgesic effects. This was achieved by fusing a BoNT/A gene to that for the light chain (LC) of type/E. The resultant purified protein, LC/E-BoNT/A, entered cultured sensory neurons and, unlike BoNT/A, inhibited release of calcitonin gene-related peptide evoked by capsaicin. Western blotting revealed that this improvement could be due to a more extensive truncation by LC/E of synaptosomal-associated protein of Mr = 25 k, essential for neuro-exocytosis. When tested in a rat spared nerve injury (SNI) model, a single intra-plantar (IPL) injection of LC/E-BoNT/A alleviated for ∼2 weeks mechanical and cold hyper-sensitivities, in a dose-dependent manner. The highest non-paralytic dose (75 U/Kg, IPL) proved significantly more efficacious than BoNT/A (15 U/Kg, IPL) or repeated systemic pregabalin (10 mg/Kg, intraperitoneal), a clinically-used pain modulator. Effects of repeated or delayed injections of this fusion protein highlighted its analgesic potential. Attenuation of mechanical hyperalgesia was extended by a second administration when the effect of the first had diminished. When injected 5 weeks after injury, LC/E-BoNT/A also reversed fully-established mechanical and cold hyper-sensitivity. Thus, combining advantageous features of BoNT/E and/A yields an efficacious, locally-applied and long-acting anti-hyperalgesic.
Collapse
Affiliation(s)
- Jiafu Wang
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Laura Casals-Diaz
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Tomas Zurawski
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Jianghui Meng
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Orla Moriarty
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - John Nealon
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Om Prakash Edupuganti
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Oliver Dolly
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
11
|
Lin M, Jiang M, Ding F, Cao Z. Syntaxin-4 and SNAP23 act as exocytic SNAREs to release NGF from cultured Schwann cells. Neurosci Lett 2017; 653:97-104. [PMID: 28119011 DOI: 10.1016/j.neulet.2017.01.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 01/14/2023]
Abstract
Nowadays peripheral nerve (PN) injury occurs more frequently, the outcome is often poor because of the ineffective treatment. Once the PN was injured, Schwann cells (SCs) release neurotrophins to guide the regeneration of axons. Recent researches revealed the duration of NGF administration acts a positive role during the nerve regeneration, but the molecular mechanisms of NGF release from SCs are unknown. To investigate components of the exocytic machinery of NGF, we used RT-PCR, Western blot and immunocytochemistry to investigate expressions and locations of soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) in rat primary cultured SCs. We found that Syntaxin-4 and SNAP23 were co-localized with NGF by immunocytochemistry. Co-immunoprecipitation (Co-IP) and RNA interference (RNAi) confirmed Syntaxin-4 associated with SNAP23 to regulate the release of NGF from SCs. Knockdown of Syntaxin-4 and SNAP23 dramatically decreased the exocytosis of NGF and inhibited the neurite outgrowth of dorsal root ganglia (DRG). Syntaxin-4 and SNAP23 acted as exocytic SNAREs to release NGF from SCs.
Collapse
Affiliation(s)
- Mengsi Lin
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China; Department of Prenatal Diagnosis, Maternal and Child Health Care Hospital of Nantong, 399 Century Avenue, Nantong, JS 226018, PR China
| | - Maorong Jiang
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China; Laboratory Animals Center, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Zheng Cao
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States.
| |
Collapse
|
12
|
Gao J, Hirata M, Mizokami A, Zhao J, Takahashi I, Takeuchi H, Hirata M. Differential role of SNAP-25 phosphorylation by protein kinases A and C in the regulation of SNARE complex formation and exocytosis in PC12 cells. Cell Signal 2015; 28:425-437. [PMID: 26721188 DOI: 10.1016/j.cellsig.2015.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/15/2015] [Accepted: 12/21/2015] [Indexed: 11/30/2022]
Abstract
The final step of regulated exocytosis, membrane fusion, is mediated by formation of the SNARE complex by syntaxin, SNAP-25 (synaptosomal-associated protein of 25 kDa), and VAMP (vesicle-associated membrane protein). Phosphorylation of SNARE and accessory proteins contributes to regulation of exocytosis. We previously identified residues of SNAP-25 phosphorylated by protein kinase A (PKA) and PKC. However, the physiological role of SNAP-25 phosphorylation in exocytosis, in particular with regard to SNARE complex formation, has remained elusive. SNARE complex formation by purified recombinant SNAP-25, syntaxin-1, and VAMP-2 in vitro was inhibited or promoted as a result of the phosphorylation at Thr(138) by PKA or at Ser(187) by PKC, respectively. SNARE complex formation in intact PC12 cells was similarly inhibited by forskolin (activator of PKA) and promoted by phorbol 12-myristate 13-acetate (PMA, activator of PKC). Noradrenaline secretion from PC12 cells induced by a high K(+) concentration was enhanced by forskolin or PMA. Stable depletion of SNAP-25 inhibited high-K(+)-induced noradrenaline secretion. Forced expression of WT SNAP-25 restored the secretory response of the SNAP-25-depleted cells to high-K(+), and this response was enhanced by forskolin or PMA. Expression of the nonphosphorylatable T138A or S187A mutants of SNAP-25 similarly rescued the secretory response to high-K(+), but the augmentation of this response by forskolin was more pronounced in the cells expressing SNAP-25 (T138A) than in those expressing SNAP-25 (WT), whereas that by PMA was less pronounced in those expressing SNAP-25 (S187A). Our results thus suggest that SNAP-25 phosphorylation by PKA or PKC contributes differentially to the control of exocytosis in PC12 cells by regulating SNARE complex formation.
Collapse
Affiliation(s)
- Jing Gao
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Makiko Hirata
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Section of Orthodontics, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiko Mizokami
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jin Zhao
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ichiro Takahashi
- Section of Orthodontics, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiroshi Takeuchi
- Division of Applied Pharmacology, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu 803-8580, Japan
| | - Masato Hirata
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
13
|
Kunieda K, Tsutsuki H, Ida T, Kishimoto Y, Kasamatsu S, Sawa T, Goshima N, Itakura M, Takahashi M, Akaike T, Ihara H. 8-Nitro-cGMP Enhances SNARE Complex Formation through S-Guanylation of Cys90 in SNAP25. ACS Chem Neurosci 2015. [PMID: 26221773 DOI: 10.1021/acschemneuro.5b00196] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nitrated guanine nucleotide 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) generated by reactive oxygen/nitrogen species causes protein S-guanylation. However, the mechanism of 8-nitro-cGMP formation and its protein targets in the normal brain have not been identified. Here, we investigated 8-nitro-cGMP generation and protein S-guanylation in the rodent brain. Immunohistochemistry indicated that 8-nitro-cGMP was produced by neurons, such as pyramidal cells and interneurons. Using liquid chromatography-tandem mass spectrometry, we determined endogenous 8-nitro-cGMP levels in the brain as 2.92 ± 0.10 pmol/mg protein. Based on S-guanylation proteomics, we identified several S-guanylated neuronal proteins, including SNAP25 which is a core member of the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) complex. SNAP25 post-translational modification including palmitoylation, phosphorylation, and oxidation, are known to regulate neurotransmission. Our results demonstrate that S-guanylation of SNAP25 enhanced the stability of the SNARE complex, which was further promoted by Ca(2+)-dependent activation of neuronal nitric oxide synthase. Using site-directed mutagenesis, we identified SNAP25 cysteine 90 as the main target of S-guanylation which enhanced the stability of the SNARE complex. The present study revealed a novel target of redox signaling via protein S-guanylation in the nervous system and provided the first substantial evidence of 8-nitro-cGMP function in the nervous system.
Collapse
Affiliation(s)
- Kohei Kunieda
- Department
of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka 599-8531, Japan
| | - Hiroyasu Tsutsuki
- Department
of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tomoaki Ida
- Department
of Environmental Health Sciences and Molecular Toxicology, Graduate
School of Medicine, Tohoku University, Miyagi 980-8575, Japan
| | - Yusuke Kishimoto
- Department
of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka 599-8531, Japan
| | - Shingo Kasamatsu
- Department
of Environmental Health Sciences and Molecular Toxicology, Graduate
School of Medicine, Tohoku University, Miyagi 980-8575, Japan
| | - Tomohiro Sawa
- Department
of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Naoki Goshima
- Quantitative
Proteomics Team, Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Makoto Itakura
- Department
of Biochemistry, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Masami Takahashi
- Department
of Biochemistry, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Takaaki Akaike
- Department
of Environmental Health Sciences and Molecular Toxicology, Graduate
School of Medicine, Tohoku University, Miyagi 980-8575, Japan
| | - Hideshi Ihara
- Department
of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka 599-8531, Japan
| |
Collapse
|
14
|
Kiris E, Nuss JE, Stanford SM, Wanner LM, Cazares L, Maestre MF, Du HT, Gomba GY, Burnett JC, Gussio R, Bottini N, Panchal RG, Kane CD, Tessarollo L, Bavari S. Phosphatase Inhibitors Function as Novel, Broad Spectrum Botulinum Neurotoxin Antagonists in Mouse and Human Embryonic Stem Cell-Derived Motor Neuron-Based Assays. PLoS One 2015; 10:e0129264. [PMID: 26061731 PMCID: PMC4462581 DOI: 10.1371/journal.pone.0129264] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/06/2015] [Indexed: 12/05/2022] Open
Abstract
There is an urgent need to develop novel treatments to counter Botulinum neurotoxin (BoNT) poisoning. Currently, the majority of BoNT drug development efforts focus on directly inhibiting the proteolytic components of BoNT, i.e. light chains (LC). Although this is a rational approach, previous research has shown that LCs are extremely difficult drug targets and that inhibiting multi-serotype BoNTs with a single LC inhibitor may not be feasible. An alternative approach would target neuronal pathways involved in intoxication/recovery, rather than the LC itself. Phosphorylation-related mechanisms have been implicated in the intoxication pathway(s) of BoNTs. However, the effects of phosphatase inhibitors upon BoNT activity in the physiological target of BoNTs, i.e. motor neurons, have not been investigated. In this study, a small library of phosphatase inhibitors was screened for BoNT antagonism in the context of mouse embryonic stem cell-derived motor neurons (ES-MNs). Four inhibitors were found to function as BoNT/A antagonists. Subsequently, we confirmed that these inhibitors protect against BoNT/A in a dose-dependent manner in human ES-MNs. Additionally, these compounds provide protection when administered in post-intoxication scenario. Importantly, the inhibitors were also effective against BoNT serotypes B and E. To the best of our knowledge, this is the first study showing phosphatase inhibitors as broad-spectrum BoNT antagonists.
Collapse
Affiliation(s)
- Erkan Kiris
- Geneva Foundation, Tacoma, WA, United States of America
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI), Frederick, MD, United States of America
| | - Jonathan E. Nuss
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Stephanie M. Stanford
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Laura M. Wanner
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Lisa Cazares
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Michael F. Maestre
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Hao T. Du
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Glenn Y. Gomba
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - James C. Burnett
- Leidos Biomedical Research, Inc., Computational Drug Development Group (CDDG), NCI, Frederick, MD, United States of America
- CDDG, Developmental Therapeutics Program, NCI, Frederick, MD, United States of America
| | - Rick Gussio
- CDDG, Developmental Therapeutics Program, NCI, Frederick, MD, United States of America
| | - Nunzio Bottini
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Rekha G. Panchal
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| | - Christopher D. Kane
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
- DoD Biotechnology High Performance Computing Software Applications Institute (BHSAI), Telemedicine and Advanced Technology Research Center (TATRC), US Army Medical Research and Materiel Command (USAMRMC), Frederick, MD, United States of America
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI), Frederick, MD, United States of America
| | - Sina Bavari
- Department of Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States of America
| |
Collapse
|
15
|
Regulation of Golgi signaling and trafficking by the KDEL receptor. Histochem Cell Biol 2013; 140:395-405. [DOI: 10.1007/s00418-013-1130-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2013] [Indexed: 12/31/2022]
|
16
|
Involvement of gecko SNAP25b in spinal cord regeneration by promoting outgrowth and elongation of neurites. Int J Biochem Cell Biol 2012; 44:2288-98. [DOI: 10.1016/j.biocel.2012.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/12/2012] [Accepted: 09/14/2012] [Indexed: 01/17/2023]
|
17
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
18
|
Hirano AA, Brandstätter JH, Morgans CW, Brecha NC. SNAP25 expression in mammalian retinal horizontal cells. J Comp Neurol 2011; 519:972-88. [PMID: 21280047 DOI: 10.1002/cne.22562] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Horizontal cells mediate inhibitory feedforward and feedback lateral interactions in the outer retina at photoreceptor terminals and bipolar cell dendrites; however, the mechanisms that underlie synaptic transmission from mammalian horizontal cells are poorly understood. The localization of a vesicular γ-aminobutyric acid (GABA) transporter (VGAT) to horizontal cell processes in primate and rodent retinae suggested that mammalian horizontal cells release transmitter in a vesicular manner. Toward determining whether the molecular machinery for vesicular transmitter release is present in horizontal cells, we investigated the expression of SNAP25 (synaptosomal-associated protein of 25 kDa), a key SNARE protein, by immunocytochemistry with cell type-specific markers in the retinae of mouse, rat, rabbit, and monkey. Different commercial antibodies to SNAP25 were tested on vertical sections of retina. We report the robust expression of SNAP25 in both plexiform layers. Double labeling with SNAP25 and calbindin antibodies demonstrated that horizontal cell processes and their endings in photoreceptor triad synapses were strongly labeled for both proteins in mouse, rat, rabbit, and monkey retinae. Double labeling with parvalbumin antibodies in monkey retina verified SNAP25 immunoreactivity in all horizontal cells. Pre-embedding immunoelectron microscopy in rabbit retina confirmed expression of SNAP25 in lateral elements within photoreceptor triad synapses. The SNAP25 immunoreactivity in the plexiform layers and outer nuclear layer fell into at least three patterns depending on the antibody, suggesting a differential distribution of SNAP25 isoforms. The presence of SNAP25a and SNAP25b isoforms in mouse retina was established by reverse transcriptase-polymerase chain reaction. SNAP25 expression in mammalian horizontal cells along with other SNARE proteins is consistent with vesicular exocytosis.
Collapse
Affiliation(s)
- Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | |
Collapse
|
19
|
Trost M, Bridon G, Desjardins M, Thibault P. Subcellular phosphoproteomics. MASS SPECTROMETRY REVIEWS 2010; 29:962-90. [PMID: 20931658 DOI: 10.1002/mas.20297] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Protein phosphorylation represents one of the most extensively studied post-translational modifications, primarily due to the emergence of sensitive methods enabling the detection of this modification both in vitro and in vivo. The availability of enrichment methods combined with sensitive mass spectrometry instrumentation has played a crucial role in uncovering the dynamic changes and the large expanding repertoire of this reversible modification. The structural changes imparted by the phosphorylation of specific residues afford exquisite mechanisms for the regulation of protein functions by modulating new binding sites on scaffold proteins or by abrogating protein-protein interactions. However, the dynamic interplay of protein phosphorylation is not occurring randomly within the cell but is rather finely orchestrated by specific kinases and phosphatases that are unevenly distributed across subcellular compartments. This spatial separation not only regulates protein phosphorylation but can also control the activity of other enzymes and the transfer of other post-translational modifications. While numerous large-scale phosphoproteomics studies highlighted the extent and diversity of phosphoproteins present in total cell lysates, the further understanding of their regulation and biological activities require a spatio-temporal resolution only achievable through subcellular fractionation. This review presents a first account of the emerging field of subcellular phosphoproteomics where cell fractionation approaches are combined with sensitive mass spectrometry methods to facilitate the identification of low abundance proteins and to unravel the intricate regulation of protein phosphorylation.
Collapse
Affiliation(s)
- Matthias Trost
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | | | | | | |
Collapse
|
20
|
Utz AC, Hirner H, Blatz A, Hillenbrand A, Schmidt B, Deppert W, Henne-Bruns D, Fischer D, Thal DR, Leithäuser F, Knippschild U. Analysis of cell type-specific expression of CK1 epsilon in various tissues of young adult BALB/c Mice and in mammary tumors of SV40 T-Ag-transgenic mice. J Histochem Cytochem 2009; 58:1-15. [PMID: 19755715 DOI: 10.1369/jhc.2009.954628] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Casein kinase 1 epsilon (CK1epsilon) is involved in various cellular processes, including cell growth, differentiation, and apoptosis, vesicle transport, and control of the circadian rhythm. Deregulation of CK1epsilon has been linked to neurodegenerative diseases and cancer. To better understand the cell type-specific functions of CK1epsilon, we determined its localization by immunhistochemistry in tissues of healthy, young adult BALB/c mice and in mammary tumors of SV40 T-antigen-transgenic mice. CK1epsilon expression was found to be highly regulated in normal tissues of endodermal, mesodermal, and ectodermal origin and in neoplastic tissue of mammary cancer. The data presented here give an overview of CK1epsilon reactivity in different organs under normal conditions and outline changes in its expression in mammary carcinomas. Our data suggest a cell/organ type-specific function of CK1epsilon and indicate that tumorigenic conversion of mammary glands in SV40 T-antigen-transgenic mice leads to downregulation of CK1epsilon. This manuscript contains online supplemental material at http://www.jhc.org. Please visit this article online to view these materials.
Collapse
Affiliation(s)
- Anja C Utz
- Department of General, Visceral, and Transplantation Surgery, University of Ulm, Steinhövelstr. 9, 89075 Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kataoka M, Sekiguchi M, Takahashi M. Identification of a minimal segment of complexin II essential for preferential distribution in axons. J Neurochem 2009; 108:1109-15. [PMID: 19141077 DOI: 10.1111/j.1471-4159.2009.05874.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Complexin II (CPLX2) is a soluble pre-synaptic protein believed to regulate neurotransmitter release from pre-synaptic terminals. CPLX2 is localized in pre-synaptic terminals in mature brain, but the mechanism of selective localization remains unclear. Here we identified an essential segment of CPLX2 for preferential axonal distribution. Myc-tagged CPLX2 was expressed in cultured rat hippocampal neurons and its distribution between axons and dendrites was compared by immunocytochemistry and image analysis. Fluorescence signals were detected in both axons and dendrites; however, their respective distribution varied significantly. Despite the fact that signal intensity decreased almost linearly from the base to the tip of the dendrite, a substantial level was sustained along the axon, even at a position near the tip. Image analyses using a series of mutants indicated that the deletion of 19 amino acid residues, G71-P89, within the 'central core' for binding to soluble N-ethylmaleimide sensitive factor attachment protein receptor proteins resulted in the loss of preferential axonal distribution. The enhanced green fluorescent protein derivative fused with the G71-P89 fragment exhibited a similar localization to that of wild type CPLX2, indicating that the G71-P89 region of CPLX2 is essential and sufficient for preferential axonal distribution.
Collapse
Affiliation(s)
- Masakazu Kataoka
- Department of Environmental Science and Technology, Shinshu University, Nagano, Japan.
| | | | | |
Collapse
|
22
|
Löhler J, Hirner H, Schmidt B, Kramer K, Fischer D, Thal DR, Leithäuser F, Knippschild U. Immunohistochemical characterisation of cell-type specific expression of CK1delta in various tissues of young adult BALB/c mice. PLoS One 2009; 4:e4174. [PMID: 19137063 PMCID: PMC2613528 DOI: 10.1371/journal.pone.0004174] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 11/24/2008] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Casein kinase 1 delta (CK1delta) phosphorylates many key proteins playing important roles in such biological processes as cell growth, differentiation, apoptosis, circadian rhythm and vesicle transport. Furthermore, deregulation of CK1delta has been linked to neurodegenerative diseases and cancer. In this study, the cell specific distribution of CK1delta in various tissues and organs of young adult BALB/c mice was analysed by immunohistochemistry. METHODOLOGY/PRINCIPAL FINDINGS Immunohistochemical staining of CK1delta was performed using three different antibodies against CK1delta. A high expression of CK1delta was found in a variety of tissues and organ systems and in several cell types of endodermal, mesodermal and ectodermal origin. CONCLUSIONS These results give an overview of the cell-type specific expression of CK1delta in different organs under normal conditions. Thus, they provide evidence for possible cell-type specific functions of CK1delta, where CK1delta can interact with and modulate the activity of key regulator proteins by site directed phosphorylation. Furthermore, they provide the basis for future analyses of CK1delta in these tissues.
Collapse
Affiliation(s)
- Jürgen Löhler
- Molecular Pathology Group, Heinrich-Pette-Institute for Experimental Immunology and Virology, University Hamburg, Hamburg, Germany
| | - Heidrun Hirner
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Bernhard Schmidt
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Klaus Kramer
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Dietmar Fischer
- Department of Experimental Neurology, University of Ulm, Ulm, Germany
| | - Dietmar R. Thal
- Laboratory of Neuropathology, Institute of Pathology, University of Ulm, Ulm, Germany
| | | | - Uwe Knippschild
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
23
|
The role of the t-SNARE SNAP-25 in action potential-dependent calcium signaling and expression in GABAergic and glutamatergic neurons. BMC Neurosci 2008; 9:105. [PMID: 18959796 PMCID: PMC2600647 DOI: 10.1186/1471-2202-9-105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 10/29/2008] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, comprised of SNAP-25, syntaxin 1A, and VAMP-2, has been shown to be responsible for action potential (AP)-dependent, calcium-triggered release of several neurotransmitters. However, this basic fusogenic protein complex may be further specialized to suit the requirements for different neurotransmitter systems, as exemplified by neurons and neuroendocrine cells. In this study, we investigate the effects of SNAP-25 ablation on spontaneous neuronal activity and the expression of functionally distinct isoforms of this t-SNARE in GABAergic and glutamatergic neurons of the adult brain. RESULTS We found that neurons cultured from Snap25 homozygous null mutant (Snap25-/-) mice failed to develop synchronous network activity seen as spontaneous AP-dependent calcium oscillations and were unable to trigger glial transients following depolarization. Voltage-gated calcium channel (VGCC) mediated calcium transients evoked by depolarization, nevertheless, did not differ between soma of SNAP-25 deficient and control neurons. Furthermore, we observed that although the expression of SNAP-25 RNA transcripts varied among neuronal populations in adult brain, the relative ratio of the transcripts encoding alternatively spliced SNAP-25 variant isoforms was not different in GABAergic and glutamatergic neurons. CONCLUSION We propose that the SNAP-25b isoform is predominantly expressed by both mature glutamatergic and GABAergic neurons and serves as a fundamental component of SNARE complex used for fast synaptic communication in excitatory and inhibitory circuits required for brain function. Moreover, SNAP-25 is required for neurons to establish AP-evoked synchronous network activity, as measured by calcium transients, whereas the loss of this t-SNARE does not affect voltage-dependent calcium entry.
Collapse
|
24
|
Auld DS, Mennicken F, Day JC, Quirion R. Neurotrophins differentially enhance acetylcholine release, acetylcholine content and choline acetyltransferase activity in basal forebrain neurons. J Neurochem 2008. [DOI: 10.1046/j.1471-4159.2001.00234.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Phosphorylation of SNAP-25 at Ser187 mediates enhancement of exocytosis by a phorbol ester in INS-1 cells. J Neurosci 2008; 28:21-30. [PMID: 18171919 DOI: 10.1523/jneurosci.2352-07.2008] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Activation of diacylglycerol (DAG) signaling pathways with phorbol esters dramatically enhances Ca2+-triggered exocytosis from both endocrine cells and neurons, however the relevant targets of DAG are controversial. A possible effector mechanism for this signaling pathway is phosphorylation of SNAP-25 (25 kDa synaptosome-associated protein) at Ser187 by PKC. Here, we investigated the role of Ser187 in the enhancement of exocytosis by the phorbol ester PMA (phorbol 12-myristate 13-acetate). We used patch-clamp measurements of membrane capacitance together with photorelease of caged-Ca2+ and membrane depolarization to study exocytosis. Expression of the nonphosphorylatable S187C SNAP-25 mutant did not attenuate the enhancement of exocytosis by PMA in either bovine chromaffin cells or the INS-1 insulin-secreting cell line. To test the effects of Ser187 mutations under conditions in which the endogenous SNAP-25 is disabled, we expressed botulinum toxin serotype E to cleave SNAP-25 in INS-1 cells. Coexpression of a toxin-resistant mutant (TR), but not wild-type SNAP-25, was able to rescue PMA-modulated exocytosis. Coexpression of the toxin with the TR-S187C SNAP-25 mutant was able to completely block the enhancement of exocytosis by PMA in response to photoelevation of [Ca2+]i to low microM levels or to a depolarizing train. The phospho-mimetic S187E mutation enhanced the small, fast burst of exocytosis evoked by photelevation of Ca2+, but, like PMA, had smaller effects on exocytosis evoked by a depolarizing train. This work supports the hypothesis that phosphorylation of Ser187 of SNAP-25 by PKC is a key step in the enhancement of exocytosis by DAG.
Collapse
|
26
|
Pozzi D, Condliffe S, Bozzi Y, Chikhladze M, Grumelli C, Proux-Gillardeaux V, Takahashi M, Franceschetti S, Verderio C, Matteoli M. Activity-dependent phosphorylation of Ser187 is required for SNAP-25-negative modulation of neuronal voltage-gated calcium channels. Proc Natl Acad Sci U S A 2008; 105:323-8. [PMID: 18162553 PMCID: PMC2224210 DOI: 10.1073/pnas.0706211105] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Indexed: 11/18/2022] Open
Abstract
Synaptosomal-associated protein of 25 kDa (SNAP-25) is a SNARE protein that regulates neurotransmission by the formation of a complex with syntaxin 1 and synaptobrevin/VAMP2. SNAP-25 also reduces neuronal calcium responses to stimuli, but neither the functional relevance nor the molecular mechanisms of this modulation have been clarified. In this study, we demonstrate that hippocampal slices from Snap25(+/-) mice display a significantly larger facilitation and that higher calcium peaks are reached after depolarization by Snap25(-/-) and Snap25(+/-) cultured neurons compared with wild type. We also show that SNAP-25b modulates calcium dynamics by inhibiting voltage-gated calcium channels (VGCCs) and that PKC phosphorylation of SNAP-25 at ser187 is essential for this process, as indicated by the use of phosphomimetic (S187E) or nonphosphorylated (S187A) mutants. Neuronal activity is the trigger that induces the transient phosphorylation of SNAP-25 at ser187. Indeed, enhancement of network activity increases the levels of phosphorylated SNAP-25, whereas network inhibition reduces the extent of protein phosphorylation. A transient peak of SNAP-25 phosphorylation also is detectable in rat hippocampus in vivo after i.p. injection with kainate to induce seizures. These findings demonstrate that differences in the expression levels of SNAP-25 impact on calcium dynamics and neuronal plasticity, and that SNAP-25 phosphorylation, by promoting inhibition of VGCCs, may mediate a negative feedback modulation of neuronal activity during intense activation.
Collapse
Affiliation(s)
- Davide Pozzi
- *Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche–Institute of Neuroscience, University of Milano, Via Vanvitelli 32, 20129 Milan, Italy
- Italian Institute of Technology, Via Morego 30, 16163 Genoa, Italy
| | - Steven Condliffe
- *Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche–Institute of Neuroscience, University of Milano, Via Vanvitelli 32, 20129 Milan, Italy
| | - Yuri Bozzi
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Via G. Moruzzi 1, 56100 Pisa, Italy
| | - Maia Chikhladze
- Istituto Neurologico C. Besta, Via Celoria 11, 20133 Milan, Italy
| | - Carlotta Grumelli
- *Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche–Institute of Neuroscience, University of Milano, Via Vanvitelli 32, 20129 Milan, Italy
| | | | - Masami Takahashi
- Department of Biochemistry, Kitasato University School of Medicine, Kanagawa 228-8555, Japan
| | | | - Claudia Verderio
- *Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche–Institute of Neuroscience, University of Milano, Via Vanvitelli 32, 20129 Milan, Italy
| | - Michela Matteoli
- *Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche–Institute of Neuroscience, University of Milano, Via Vanvitelli 32, 20129 Milan, Italy
- **Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Don C. Gnocchi, 20129 Milan, Italy; and
| |
Collapse
|
27
|
Ziegler CG, Sicard F, Lattke P, Bornstein SR, Ehrhart-Bornstein M, Krug AW. Dehydroepiandrosterone induces a neuroendocrine phenotype in nerve growth factor-stimulated chromaffin pheochromocytoma PC12 cells. Endocrinology 2008; 149:320-8. [PMID: 17884937 DOI: 10.1210/en.2007-0645] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The adrenal androgen dehydroepiandrosterone (DHEA) is produced in the inner zone of the adrenal cortex, which is in direct contact to adrenal medullary cells. Due to their close anatomical proximity and tightly intermingled cell borders, a direct interaction of adrenal cortex and medulla has been postulated. In humans congenital adrenal hyperplasia due to 21-hydroxylase deficiency results in androgen excess accompanied by severe adrenomedullary dysplasia and chromaffin cell dysfunction. Therefore, to define the mechanisms of DHEA action on chromaffin cell function, we investigated its effect on cell survival and differentiation processes on a molecular level in the chromaffin cell line PC12. DHEA lessened the positive effect of NGF on cell survival and neuronal differentiation. Nerve growth factor (NGF)-mediated induction of a neuronal phenotype was inhibited by DHEA as indicated by reduced neurite outgrowth and decreased expression of neuronal marker proteins such as synaptosome-associated protein of 25 kDa and vesicle-associated membrane protein-2. We examined whether DHEA may stimulate the cells toward a neuroendocrine phenotype. DHEA significantly elevated catecholamine release from unstimulated PC12 cells in the presence but not absence of NGF. Accordingly, DHEA enhanced the expression of the neuroendocrine marker protein chromogranin A. Next, we explored the possible molecular mechanisms of DHEA and NGF interaction. We demonstrate that NGF-induced ERK1/2 phosphorylation was reduced by DHEA. In summary, our data show that DHEA influences cell survival and differentiation processes in PC12 cells, possibly by interacting with the ERK1/2 MAPK pathway. DHEA drives NGF-stimulated cells toward a neuroendocrine phenotype, suggesting that the interaction of intraadrenal steroids and growth factors is required for the maintenance of an intact adrenal medulla.
Collapse
Affiliation(s)
- Christian G Ziegler
- University Hospital Carl Gustav Carus, Medical Clinic III, University of Dresden, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
G-proteins (guanine nucleotide-binding proteins) are membrane-attached proteins composed of three subunits, alpha, beta, and gamma. They transduce signals from G-protein coupled receptors (GPCRs) to target effector proteins. The agonistactivated receptor induces a conformational change in the G-protein trimer so that the alpha-subunit binds GTP in exchange for GDP and alpha-GTP, and betagamma-subunits separate to interact with the target effector. Effector-interaction is terminated by the alpha-subunit GTPase activity, whereby bound GTP is hydrolyzed to GDP. This is accelerated in situ by RGS proteins, acting as GTPase-activating proteins (GAPs). Galpha-GDP and Gbetagamma then reassociate to form the Galphabetagamma trimer. G-proteins primarily involved in the modulation of neurotransmitter release are G(o), G(q) and G(s). G(o) mediates the widespread presynaptic auto-inhibitory effect of many neurotransmitters (e.g., via M2/M4 muscarinic receptors, alpha(2) adrenoreceptors, micro/delta opioid receptors, GABAB receptors). The G(o) betagamma-subunit acts in two ways: first, and most ubiquitously, by direct binding to CaV2 Ca(2+) channels, resulting in a reduced sensitivity to membrane depolarization and reduced Ca(2+) influx during the terminal action potential; and second, through a direct inhibitory effect on the transmitter release machinery, by binding to proteins of the SNARE complex. G(s) and G(q) are mainly responsible for receptor-mediated facilitatory effects, through activation of target enzymes (adenylate cyclase, AC and phospholipase-C, PLC respectively) by the GTP-bound alpha-subunits.
Collapse
Affiliation(s)
- David A Brown
- Department of Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| | | |
Collapse
|
29
|
Cunha DA, Roma LP, Boschero AC. Prolactin modulates the association and phosphorylation of SNARE and kinesin/MAP-2 proteins in neonatal pancreatic rat islets. Mol Cell Endocrinol 2007; 273:32-41. [PMID: 17573185 DOI: 10.1016/j.mce.2007.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 05/02/2007] [Accepted: 05/08/2007] [Indexed: 11/18/2022]
Abstract
Prolactin induces maturation of insulin secretion in cultured neonatal rat islets. In this study, we investigated whether the improved secretory response to glucose caused by prolactin involves alteration in the expression, association and phosphorylation of several proteins that participate in these processes. Messenger RNA was extracted from neonatal rat islets cultured for 5 days in the presence of prolactin and reverse transcribed. Gene expression was analyzed by semi-quantitative RT-PCR and by Western blotting for proteins. The gene transcription and protein expression of kinesin and MAP-2 were increased in prolactin-treated islets compared to the controls. The association and phosphorylation of proteins was analyzed by immunoprecipitation followed by Western blotting, after acute exposure to prolactin. Prolactin increased the association between SNARE proteins and kinesin/MAP-2 while the association of munc-18/syntaxin 1A was decreased. Serine phosphorylation of SNAP-25, syntaxin 1A, munc-18, MAP-2 was significantly higher whereas kinesin phosphorylation was decreased in prolactin-treated islets. There was an increase in SNARE complex formation in islets stimulated with prolactin, 22 mM glucose, 40 mM K(+), 200 microM carbachol and 1 microM PMA. The prolactin-induced increase in the formation of SNARE complex and syntaxin 1A phosphorylation was inhibited by PD098059 and U0126, inhibitors of the MAPK pathway. These findings indicate that prolactin primes pancreatic beta-cells to release insulin by increasing the expression and phosphorylation/association of proteins implicated in the secretory machinery and the MAPK/PKC pathway is important for this effect.
Collapse
Affiliation(s)
- Daniel A Cunha
- Department of Physiology and Biophysics, Institute of Biology, State University of Campinas (UNICAMP), CP 6109, Campinas 13083-970, SP, Brazil
| | | | | |
Collapse
|
30
|
Yang Y, Craig TJ, Chen X, Ciufo LF, Takahashi M, Morgan A, Gillis KD. Phosphomimetic mutation of Ser-187 of SNAP-25 increases both syntaxin binding and highly Ca2+-sensitive exocytosis. ACTA ACUST UNITED AC 2007; 129:233-44. [PMID: 17325194 PMCID: PMC2151612 DOI: 10.1085/jgp.200609685] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The phosphorylation targets that mediate the enhancement of exocytosis by PKC are unknown. PKC phosporylates the SNARE protein SNAP-25 at Ser-187. We expressed mutants of SNAP-25 using the Semliki Forest Virus system in bovine adrenal chromaffin cells and then directly measured the Ca2+ dependence of exocytosis using photorelease of caged Ca2+ together with patch-clamp capacitance measurements. A flash of UV light used to elevate [Ca2+]i to several μM and release the highly Ca2+-sensitive pool (HCSP) of vesicles was followed by a train of depolarizing pulses to elicit exocytosis from the less Ca2+-sensitive readily releasable pool (RRP) of vesicles. Carbon fiber amperometry confirmed that the amount and kinetics of catecholamine release from individual granules were similar for the two phases of exocytosis. Mimicking PKC phosphorylation with expression of the S187E SNAP-25 mutant resulted in an approximately threefold increase in the HCSP, whereas the response to depolarization increased only 1.5-fold. The phosphomimetic S187D mutation resulted in an ∼1.5-fold increase in the HCSP but a 30% smaller response to depolarization. In vitro binding assays with recombinant SNARE proteins were performed to examine shifts in protein–protein binding that may promote the highly Ca2+-sensitive state. The S187E mutant exhibited increased binding to syntaxin but decreased Ca2+-independent binding to synaptotagmin I. Mimicking phosphorylation of the putative PKA phosphorylation site of SNAP-25 with the T138E mutation decreased binding to both syntaxin and synaptotagmin I in vitro. Expressing the T138E/ S187E double mutant in chromaffin cells demonstrated that enhancing the size of the HCSP correlates with an increase in SNAP-25 binding to syntaxin in vitro, but not with Ca2+-independent binding of SNAP-25 to synaptotagmin I. Our results support the hypothesis that exocytosis triggered by lower Ca2+ concentrations (from the HCSP) occurs by different molecular mechanisms than exocytosis triggered by higher Ca2+ levels.
Collapse
Affiliation(s)
- Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Fritzius T, Frey AD, Schweneker M, Mayer D, Moelling K. WD-repeat-propeller-FYVE protein, ProF, binds VAMP2 and protein kinase Czeta. FEBS J 2007; 274:1552-66. [PMID: 17313651 DOI: 10.1111/j.1742-4658.2007.05702.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have recently identified a protein, consisting of seven WD repeats, presumably forming a beta-propeller, and a domain identified in Fab1p, YOTB, VAC1p, and EEA1 (FYVE) domain, ProF. The FYVE domain targets the protein to vesicular membranes, while the WD repeats allow binding of the activated kinases Akt and protein kinase (PK)Czeta. Here, we describe the vesicle-associated membrane protein 2 (VAMP2) as interaction partner of ProF. The interaction is demonstrated with overexpressed and endogenous proteins in mammalian cells. ProF and VAMP2 partially colocalize on vesicular structures with PKCzeta and the proteins form a ternary complex. VAMP2 can be phosphorylated by activated PKCzeta in vitro and the presence of ProF increases the PKCzeta-dependent phosphorylation of VAMP2 in vitro. ProF is an adaptor protein that brings together a kinase with its substrate. VAMP2 is known to regulate docking and fusion of vesicles and to play a role in targeting vesicles to the plasma membrane. The complex may be involved in vesicle cycling in various secretory pathways.
Collapse
Affiliation(s)
- Thorsten Fritzius
- Institute of Medical Virology, University of Zurich, Gloriastrasse 30, Zurich CH-8006, Switzerland
| | | | | | | | | |
Collapse
|
32
|
Wolff S, Stöter M, Giamas G, Piesche M, Henne-Bruns D, Banting G, Knippschild U. Casein kinase 1 delta (CK1delta) interacts with the SNARE associated protein snapin. FEBS Lett 2006; 580:6477-84. [PMID: 17101137 DOI: 10.1016/j.febslet.2006.10.068] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 09/21/2006] [Accepted: 10/27/2006] [Indexed: 11/21/2022]
Abstract
In this study we identified snapin as an interaction partner of the CK1 isoform delta (CK1delta) in the yeast two-hybrid system and localized the interacting domains of both proteins. The interaction of CK1delta with snapin was confirmed by co-immunoprecipitation. Snapin was phosphorylated by CK1delta in vitro. Both proteins localized in close proximity in the perinuclear region, wherein snapin was found to associate with membranes of the Golgi apparatus. The identification of snapin as a new substrate of CK1delta points towards a possible function for CK1delta in modulating snapin specific functions.
Collapse
Affiliation(s)
- Sonja Wolff
- Clinic of General-, Visceral- and Transplantation Surgery, the Medical University of Ulm, Steinhövelstr. 9, 89075 Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Kataoka M, Kuwahara R, Matsuo R, Sekiguchi M, Inokuchi K, Takahashi M. Development- and activity-dependent regulation of SNAP-25 phosphorylation in rat brain. Neurosci Lett 2006; 407:258-62. [PMID: 16978778 DOI: 10.1016/j.neulet.2006.08.055] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 08/21/2006] [Accepted: 08/22/2006] [Indexed: 11/22/2022]
Abstract
Synaptosomal-associated protein of 25kDa (SNAP-25), a member of the SNARE proteins essential for neurotransmitter release, is phosphorylated at Ser(187) in PC12 cells and in the rat brain in a protein kinase C-dependent manner. It remains unclear how the phosphorylation of SNAP-25 is regulated during development and by neuronal activity. We studied the mode of SNAP-25 phosphorylation at Ser(187) in the rat brain using an anti-phosphorylated SNAP-25 antibody. Both the expression and phosphorylation of SNAP-25 increased remarkably during the early postnatal period, but their onsets were quite different. SNAP-25 expression was detected as early as embryonic Day 18, whereas the phosphorylation of SNAP-25 could not be detected until postnatal Day 4. A delay in the onset of phosphorylation was also observed in cultured rat hippocampal neurons. The phosphorylation of SNAP-25 was regulated in a neuronal activity-dependent manner and, in the rat hippocampus, decreased by introducing seizures with kainic acid. These results clearly indicated that the phosphorylation of SNAP-25 at Ser(187) is regulated in development- and neuronal activity-dependent manners, and is likely to play important roles in higher brain functions.
Collapse
Affiliation(s)
- Masakazu Kataoka
- Department of Environmental Science and Technology, Faculty of Engineering, Shinshu University, Wakasato 4-17-1, Nagano-shi, Nagano, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Pooley RD, Reddy S, Soukoulis V, Roland JT, Goldenring JR, Bader DM. CytLEK1 is a regulator of plasma membrane recycling through its interaction with SNAP-25. Mol Biol Cell 2006; 17:3176-86. [PMID: 16672379 PMCID: PMC1483049 DOI: 10.1091/mbc.e05-12-1127] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 04/17/2006] [Accepted: 04/25/2006] [Indexed: 12/26/2022] Open
Abstract
SNAP-25 is a component of the SNARE complex that is involved in membrane docking and fusion. Using a yeast two-hybrid screen, we identify a novel interaction between SNAP-25 and cytoplasmic Lek1 (cytLEK1), a protein previously demonstrated to associate with the microtubule network. The binding domains within each protein were defined by yeast two-hybrid, coimmunoprecipitation, and colocalization studies. Confocal analyses reveal a high degree of colocalization between the proteins. In addition, the endogenous proteins can be isolated as a complex by immunoprecipitation. Further analyses demonstrate that cytLEK1 and SNAP-25 colocalize and coprecipitate with Rab11a, myosin Vb, VAMP2, and syntaxin 4, components of the plasma membrane recycling pathway. Overexpression of the SNAP-25-binding domain of cytLEK1, and depletion of endogenous Lek1 alters transferrin trafficking, consistent with a function in vesicle recycling. Taken together, our studies indicate that cytLEK1 is a link between recycling vesicles and the microtubule network through its association with SNAP-25. This interaction may play a key role in the regulation of the recycling endosome pathway.
Collapse
Affiliation(s)
- Ryan D. Pooley
- *Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300; and
| | - Samyukta Reddy
- *Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300; and
| | - Victor Soukoulis
- *Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300; and
| | - Joseph T. Roland
- Department of Surgery and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, and Nashville VAMC, Nashville, TN 37212-2175
| | - James R. Goldenring
- Department of Surgery and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, and Nashville VAMC, Nashville, TN 37212-2175
| | - David M. Bader
- *Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300; and
| |
Collapse
|
35
|
Abstract
PKC (protein kinase C) has been known for many years to modulate regulated exocytosis in a wide variety of cell types. In neurons and neuroendocrine cells, PKC regulates several different stages of the exocytotic process, suggesting that these multiple actions of PKC are mediated by phosphorylation of distinct protein targets. In recent years, a variety of exocytotic proteins have been identified as PKC substrates, the best characterized of which are SNAP-25 (25 kDa synaptosome-associated protein) and Munc18. In the present study, we review recent evidence suggesting that site-specific phosphorylation of SNAP-25 and Munc18 by PKC regulates distinct stages of exocytosis.
Collapse
|
36
|
Leenders AM, Sheng ZH. Modulation of neurotransmitter release by the second messenger-activated protein kinases: implications for presynaptic plasticity. Pharmacol Ther 2005; 105:69-84. [PMID: 15626456 PMCID: PMC1804289 DOI: 10.1016/j.pharmthera.2004.10.012] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Activity-dependent modulation of synaptic function and structure is emerging as one of the key mechanisms underlying synaptic plasticity. Whereas over the past decade considerable progress has been made in identifying postsynaptic mechanisms for synaptic plasticity, the presynaptic mechanisms involved have remained largely elusive. Recent evidence implicates that second messenger regulation of the protein interactions in synaptic vesicle release machinery is one mechanism by which cellular events modulate synaptic transmission. Thus, identifying protein kinases and their targets in nerve terminals, particularly those functionally regulated by synaptic activity or intracellular [Ca2+], is critical to the elucidation of the molecular mechanisms underlying modulation of neurotransmitter release and presynaptic plasticity. The phosphorylation and dephosphorylation states of synaptic proteins that mediate vesicle exocytosis could regulate the biochemical pathways leading from synaptic vesicle docking to fusion. However, functional evaluation of the activity-dependent phosphorylation events for modulating presynaptic functions still represents a considerable challenge. Here, we present a brief overview of the data on the newly identified candidate targets of the second messenger-activated protein kinases in the presynaptic release machinery and discuss the potential impact of these phosphorylation events in synaptic strength and presynaptic plasticity.
Collapse
Affiliation(s)
| | - Zu-Hang Sheng
- * Corresponding author. Tel.: 301 435 4596; fax: 301 480 5763. E-mail address: (Z.-H. Sheng)
| |
Collapse
|
37
|
Elbert M, Rossi G, Brennwald P. The yeast par-1 homologs kin1 and kin2 show genetic and physical interactions with components of the exocytic machinery. Mol Biol Cell 2005; 16:532-49. [PMID: 15563607 PMCID: PMC545889 DOI: 10.1091/mbc.e04-07-0549] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2004] [Revised: 10/28/2004] [Accepted: 11/11/2004] [Indexed: 11/11/2022] Open
Abstract
Kin1 and Kin2 are Saccharomyces cerevisiae counterparts of Par-1, the Caenorhabditis elegans kinase essential for the establishment of polarity in the one cell embryo. Here, we present evidence for a novel link between Kin1, Kin2, and the secretory machinery of the budding yeast. We isolated KIN1 and KIN2 as suppressors of a mutant form of Rho3, a Rho-GTPase acting in polarized trafficking. Genetic analysis suggests that KIN1 and KIN2 act downstream of the Rab-GTPase Sec4, its exchange factor Sec2, and several components of the vesicle tethering complex, the Exocyst. We show that Kin1 and Kin2 physically interact with the t-SNARE Sec9 and the Lgl homologue Sro7, proteins acting at the final stage of exocytosis. Structural analysis of Kin2 reveals that its catalytic activity is essential for its function in the secretory pathway and implicates the conserved 42-amino acid tail at the carboxy terminal of the kinase in autoinhibition. Finally, we find that Kin1 and Kin2 induce phosphorylation of t-SNARE Sec9 in vivo and stimulate its release from the plasma membrane. In summary, we report the finding that yeast Par-1 counterparts are associated with and regulate the function of the exocytic apparatus via phosphorylation of Sec9.
Collapse
Affiliation(s)
- Maya Elbert
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
38
|
Hepp R, Puri N, Hohenstein AC, Crawford GL, Whiteheart SW, Roche PA. Phosphorylation of SNAP-23 Regulates Exocytosis from Mast Cells. J Biol Chem 2005; 280:6610-20. [PMID: 15611044 DOI: 10.1074/jbc.m412126200] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulated exocytosis is a process in which a physiological trigger initiates the translocation, docking, and fusion of secretory granules with the plasma membrane. A class of proteins termed SNAREs (including SNAP-23, syntaxins, and VAMPs) are known regulators of secretory granule/plasma membrane fusion events. We have investigated the molecular mechanisms of regulated exocytosis in mast cells and find that SNAP-23 is phosphorylated when rat basophilic leukemia mast cells are triggered to degranulate. The kinetics of SNAP-23 phosphorylation mirror the kinetics of exocytosis. We have identified amino acid residues Ser(95) and Ser(120) as the major phosphorylation sites in SNAP-23 in rodent mast cells. Quantitative analysis revealed that approximately 10% of SNAP-23 was phosphorylated when mast cell degranulation was induced. These same residues were phosphorylated when mouse platelet degranulation was induced with thrombin, demonstrating that phosphorylation of SNAP-23 Ser(95) and Ser(120) is not restricted to mast cells. Although triggering exocytosis did not alter the absolute amount of SNAP-23 bound to SNAREs, after stimulation essentially all of the SNAP-23 bound to the plasma membrane SNARE syntaxin 4 and the vesicle SNARE VAMP-2 was phosphorylated. Regulated exocytosis studies revealed that overexpression of SNAP-23 phosphorylation mutants inhibited exocytosis from rat basophilic leukemia mast cells, demonstrating that phosphorylation of SNAP-23 on Ser(120) and Ser(95) modulates regulated exocytosis by mast cells.
Collapse
Affiliation(s)
- Régine Hepp
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
39
|
Rüder C, Reimer T, Delgado-Martinez I, Hermosilla R, Engelsberg A, Nehring R, Dörken B, Rehm A. EBAG9 adds a new layer of control on large dense-core vesicle exocytosis via interaction with Snapin. Mol Biol Cell 2005; 16:1245-57. [PMID: 15635093 PMCID: PMC551489 DOI: 10.1091/mbc.e04-09-0817] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Regulated exocytosis is subject to several modulatory steps that include phosphorylation events and transient protein-protein interactions. The estrogen receptor-binding fragment-associated gene9 (EBAG9) gene product was recently identified as a modulator of tumor-associated O-linked glycan expression in nonneuronal cells; however, this molecule is expressed physiologically in essentially all mammalian tissues. Particular interest has developed toward this molecule because in some human tumor entities high expression levels correlated with clinical prognosis. To gain insight into the cellular function of EBAG9, we scored for interaction partners by using the yeast two-hybrid system. Here, we demonstrate that EBAG9 interacts with Snapin, which is likely to be a modulator of Synaptotagmin-associated regulated exocytosis. Strengthening of this interaction inhibited regulated secretion of neuropeptide Y from PC12 cells, whereas evoked neurotransmitter release from hippocampal neurons remained unaltered. Mechanistically, EBAG9 decreased phosphorylation of Snapin; subsequently, association of Snapin with synaptosome-associated protein of 25 kDa (SNAP25) and SNAP23 was diminished. We suggest that the occurrence of SNAP23, Snapin, and EBAG9 also in nonneuronal cells might extend the modulatory role of EBAG9 to a broad range of secretory cells. The conjunction between EBAG9 and Snapin adds an additional layer of control on exocytosis processes; in addition, mechanistic evidence is provided that inhibition of phosphorylation has a regulatory function in exocytosis.
Collapse
Affiliation(s)
- Constantin Rüder
- Department of Hematology, Oncology, and Tumor Immunology, Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Xu NJ, Yu YX, Zhu JM, Liu H, Shen L, Zeng R, Zhang X, Pei G. Inhibition of SNAP-25 Phosphorylation at Ser187 Is Involved in Chronic Morphine-induced Down-regulation of SNARE Complex Formation. J Biol Chem 2004; 279:40601-8. [PMID: 15277518 DOI: 10.1074/jbc.m406896200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Opiate abuse has been shown to cause adaptive changes in presynaptic release and protein phosphorylation-mediated synaptic plasticity, but the underlying mechanisms remain unclear. Neuronal SNARE proteins serve as important regulatory molecules underlying neural plasticity in view of their major role in the process of neurotransmitter release. In the present study, the expression of SNAP-25, a t-SNARE protein essential for vesicle release, was found to be dramatically regulated in hippocampus after chronic morphine treatment, which was visualized with two-dimensional gel electrophoresis. The spots of SNAP-25 in the gel were shifted along the dimension of isoelectric point, indicating a likely change of the post-transcriptional modification. Immunoblotting analysis with specific antibody to Ser187, a protein kinase C (PKC) phosphorylation site of SNAP-25, revealed that the specific phosphorylation was correspondingly decreased, which was correlated with morphine-induced inhibition of PKC activity. Moreover, the level of ternary complex of SNARE proteins in either synaptosomes or PC12 cells was significantly reduced after chronic morphine treatment. This suggests a causal relationship between the inhibition of PKC-dependent SNAP-25 phosphorylation and the down-regulation of SNARE complex formation after chronic morphine treatment. Further analysis of SNARE complex formed by transfection of the wild-type or Ser187 mutants of SNAP-25 showed that only wild-type-formed complex was inhibited by morphine treatment. Thus, these results indicate that chronic morphine treatment inhibits phosphorylation of SNAP-25 at Ser187 and leads to a down-regulation of SNARE complex formation, which presents a potential molecular mechanism for the alteration of exocytotic process and neural plasticity during opiate abuse.
Collapse
Affiliation(s)
- Nan-Jie Xu
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Institute of Neuroscience, Graduate School of the Chinese Academy of Sciences, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Takahashi M, Itakura M, Kataoka M. New Aspects of Neurotransmitter Releasee and Exocytosis: Regulation of Neurotransmitter Release by Phosphorylation. J Pharmacol Sci 2003; 93:41-5. [PMID: 14501150 DOI: 10.1254/jphs.93.41] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Synaptic transmission is conducted by neurotransmitters released from nerve terminals. Neurotransmitter release is regulated both positively and negatively by multiple mechanisms, and its regulation is believed to be one of the important mechanisms of synaptic plasticity underlying learning and memory. Various protein kinases play important roles in the regulation, and candidates for protein substrates essential for the regulation have been identified.
Collapse
Affiliation(s)
- Masami Takahashi
- Department of Biochemistry, Kitasato University School of Medicine, Kanagawa, Japan.
| | | | | |
Collapse
|
42
|
Hepp R, Cabaniols JP, Roche PA. Differential phosphorylation of SNAP-25 in vivo by protein kinase C and protein kinase A. FEBS Lett 2002; 532:52-6. [PMID: 12459461 DOI: 10.1016/s0014-5793(02)03629-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
SNAP-25 is a key protein required for the fusion of synaptic vesicles with the plasma membrane during exocytosis. This study establishes that SNAP-25 is differentially phosphorylated by protein kinase C and protein kinase A in neuroendocrine PC12 cells. Using phosphopeptide mapping and site-directed mutagenesis we identified both Thr138 and Ser187 as the targets of SNAP-25 phosphorylation by protein kinase C and Thr138 as the exclusive site of SNAP-25 phosphorylation by protein kinase A in vivo. Finally, despite published data to the contrary, we demonstrate that stimulation of regulated exocytosis under physiological conditions is independent of a measurable increase in SNAP-25 phosphorylation in PC12 cells.
Collapse
Affiliation(s)
- Régine Hepp
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bldg. 10, Room 4B36, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
43
|
Protein kinase C-dependent phosphorylation of synaptosome-associated protein of 25 kDa at Ser187 potentiates vesicle recruitment. J Neurosci 2002. [PMID: 12417653 DOI: 10.1523/jneurosci.22-21-09278.2002] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Activation of protein kinase C (PKC) constitutes a key event in the upregulation of secretory strength in neurons and neurosecretory cells during extensive stimulation, presumably by speeding up vesicle supply. However, the molecular targets and their mode of action remain elusive. We studied the only PKC-dependent phosphorylation site in the neuronal soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, Ser(187), in synaptosome-associated protein of 25 kDa (SNAP-25). This phosphorylation site is located within the negatively charged C-terminal end of SNAP-25, which has been shown to be of critical importance in calcium-triggered exocytosis. We combined mutational studies that used overexpression in chromaffin cells with capacitance measurements and flash photolysis of caged calcium, allowing for high time resolution during both the stimulation and measurement of exocytosis. Overexpression of mutants simulating the phosphorylated form of Ser(187) accelerated vesicle recruitment after the emptying of the releasable vesicle pools. Overexpression of mutants simulating the nonphosphorylated form, or block of PKC, impaired the refilling of the vesicle pools to similar extents. Biochemical studies verified the phosphorylation of a subpopulation of SNAP-25 after elevation of intracellular calcium concentrations. Some of the mutations led to a moderately decreased fast exocytotic burst component, which did not seem to be associated with the phosphorylation state of SNAP-25. Thus the C terminus of SNAP-25 plays a role for both fast exocytosis triggering and vesicle recruitment, and the latter process is regulated by PKC-dependent phosphorylation.
Collapse
|
44
|
Gonelle-Gispert C, Costa M, Takahashi M, Sadoul K, Halban P. Phosphorylation of SNAP-25 on serine-187 is induced by secretagogues in insulin-secreting cells, but is not correlated with insulin secretion. Biochem J 2002; 368:223-32. [PMID: 12164783 PMCID: PMC1222969 DOI: 10.1042/bj20020896] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2002] [Revised: 07/29/2002] [Accepted: 08/06/2002] [Indexed: 11/17/2022]
Abstract
The tSNARE (the target-membrane soluble NSF-attachment protein receptor, where NSF is N -ethylmaleimide-sensitive fusion protein) synaptosomal-associated protein of 25 kDa (SNAP-25) is implicated in regulated insulin secretion. In pheochromocytoma PC12 cells, SNAP-25 is phosphorylated at Ser(187), which lies in a region that is important for its function. The aims of the present study were to determine whether SNAP-25 is phosphorylated at Ser(187) in insulin-secreting cells and, if so, whether this is important for regulated insulin secretion. The major findings are: (i) SNAP-25 is rapidly and reversibly phosphorylated on Ser(187) in both rat insulinoma INS-1 cells and rat islets in response to the phorbol ester, PMA; (ii) less than 35% of SNAP-25 in INS-1 cells is phosphorylated in response to PMA, and phosphorylation is limited to plasma-membrane-associated SNAP-25; (iii) both SNAP-25 isoforms (a and b) are phosphorylated, with 1.8-fold greater phosphorylation for SNAP-25b in response to PMA; (iv) in rat islets, Ser(187) phosphorylation is stimulated by glucose or carbachol, albeit to a lesser extent than by PMA, but not by cAMP; (v) insulin secretion from botulinum neurotoxin E-treated hamster insulinoma tumour (HIT) cells, transfected with toxin-resistant Ser(187)-->Ala or Ser(187)-->Asp mutant SNAP-25, was similar to that of wild-type HIT cells. Furthermore, in rat islets no correlation was found between the extent of SNAP-25 phosphorylation at Ser(187) in response to secretagogues and stimulation of insulin release; (vi) use of protein kinase C (PKC) inhibitors suggests that glucose stimulates SNAP-25 phosphorylation via conventional and non-conventional PKC isoforms. In summary, although SNAP-25 phosphorylation at Ser(187) occurs in insulin-secreting cells and is mediated by PKC, it does not appear to play a major role in regulated insulin secretion.
Collapse
Affiliation(s)
- Carmen Gonelle-Gispert
- Laboratoires de Recherche Louis Jeantet, Centre Médical Universitaire, rue Michel Servet, CH-1211 Geneva 4, Switzerland.
| | | | | | | | | |
Collapse
|
45
|
Abstract
Many neurons release a variety of amino acids in response to depolarizing stimuli. Although some of these amino acids, namely, glutamate, aspartate, and gamma-aminobutyric acid (GABA), have been qualified as neurotransmitters, functional roles of the other amino acids including alanine remain obscure. We investigated the mechanism and the origin of alanine release from cultured rat cerebellar cells. High-K(+)-induced depolarization produced a considerable amount (139+/-8 pmol/2 min/dish) of alanine release, comparable to that of glutamate (103+/-7 pmol/2 min/dish). Other depolarizing agents including veratridine or 4-aminopyridine also induced alanine release, suggesting that the major source is excitable neurons, rather than non-excitable glial cells. Depolarization-evoked alanine release was suppressed in the absence of extracellular Ca(2+), and was almost abolished by treating the cells with botulinum type B neurotoxin (BoNT/B), indicating that alanine is released by Ca(2+)-dependent exocytosis of vesicle-associated membrane protein-2 (VAMP-2)-containing vesicles. The properties of alanine release were different from those of glutamate and GABA in several aspects: (a) Depolarization-dependent alanine release appeared as early as 7 days in vitro, much earlier than that of GABA. (b) Fifty microM kainate, which causes selective cell death of GABAergic neurons in the culture, only partially reduced alanine release, whereas it had no effect on glutamate release. (c) Alanine release was not affected by phorbol ester, which enhanced glutamate and GABA release in a kinase-dependent manner. We therefore conclude that alanine release occurs via exocytosis of a pool of synaptic vesicles distinct from those containing glutamate or GABA.
Collapse
Affiliation(s)
- Takeshi Koga
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, University of Tokyo, Meguro-ku, Tokyo 153-8902, Japan
| | | | | |
Collapse
|
46
|
Sombers LA, Colliver TL, Ewing AG. Differentiated PC12 cells: a better model system for the study of the VMAT's effects on neuronal communication. Ann N Y Acad Sci 2002; 971:86-8. [PMID: 12438095 DOI: 10.1111/j.1749-6632.2002.tb04439.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Leslie A Sombers
- Department of Chemistry, 152 Davey Laboratory, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | |
Collapse
|
47
|
Gurunathan S, Marash M, Weinberger A, Gerst JE. t-SNARE phosphorylation regulates endocytosis in yeast. Mol Biol Cell 2002; 13:1594-607. [PMID: 12006655 PMCID: PMC111129 DOI: 10.1091/mbc.01-11-0541] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Earlier we demonstrated that activation of a ceramide-activated protein phosphatase (CAPP) conferred normal growth and secretion to yeast lacking their complement of exocytic v-SNAREs (Snc1,2) or bearing a temperature-sensitive mutation in an exocytic t-SNARE (Sso2). CAPP activation led to Sso dephosphorylation and enhanced the assembly of t-SNAREs into functional complexes. Thus, exocytosis in yeast is modulated by t-SNARE phosphorylation. Here, we show that endocytic defects in cells lacking the v- and t-SNAREs involved in endocytosis are also rescued by CAPP activation. Yeast lacking the Tlg1 or Tlg2 t-SNAREs, the Snc v-SNAREs, or both, undergo endocytosis after phosphatase activation. CAPP activation correlated with restored uptake of FM4-64 to the vacuole, the uptake and degradation of the Ste2 receptor after mating factor treatment, and the dephosphorylation and assembly of Tlg1,2 into SNARE complexes. Activation of the phosphatase by treatment with C(2)-ceramide, VBM/ELO gene inactivation, or by the overexpression of SIT4 was sufficient to confer rescue. Finally, we found that mutation of single PKA sites in Tlg1 (Ser31 to Ala31) or Tlg2 (Ser90 to Ala90) was sufficient to restore endocytosis, but not exocytosis, to snc cells. These results suggest that endocytosis is also modulated by t-SNARE phosphorylation in vivo.
Collapse
|
48
|
Shoji-Kasai Y, Itakura M, Kataoka M, Yamamori S, Takahashi M. Protein kinase C-mediated translocation of secretory vesicles to plasma membrane and enhancement of neurotransmitter release from PC12 cells. Eur J Neurosci 2002; 15:1390-4. [PMID: 11994133 DOI: 10.1046/j.1460-9568.2002.01972.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In order to elucidate the molecular mechanism of phorbol ester-induced potentiation of neurotransmitter release, changes in the subcellular distribution of secretory vesicles were studied in PC12 cells. Dopamine (DA) and acetylcholine containing vesicles were selectively labelled by expressing green fluorescent protein-conjugated vesicular monoamine transporter and vesicular acetylcholine transporter, respectively. In the resting state, these vesicles were distributed throughout the cytoplasm. Phorbol-12-myristate-13-acetate (PMA), but not the inactive analogue 4 alpha-PMA, induced a redistribution of both types of secretory vesicles near the plasma membrane, and this change was abolished by a protein kinase C (PKC) inhibitor, bisindolylmaleimide I (BIS). PMA also induced a marked enhancement of depolarization-induced DA release and phosphorylation of SNAP-25 at Ser187. BIS completely inhibited PMA-induced SNAP-25 phosphorylation but suppressed PMA-induced enhancement of DA release only partially. These results suggest that PMA enhances neurotransmitter release from PC12 cells by both PKC-dependent and PKC-independent mechanisms, and PKC enhances neurotransmitter release by recruiting secretory vesicles to the plasma membrane.
Collapse
Affiliation(s)
- Yoko Shoji-Kasai
- Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan
| | | | | | | | | |
Collapse
|