1
|
Ma L, Sun D, Wen S, Yuan J, Li J, Tan X, Cao S. PSD-95 Protein: A Promising Therapeutic Target in Chronic Pain. Mol Neurobiol 2025; 62:3361-3375. [PMID: 39285025 DOI: 10.1007/s12035-024-04485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 09/04/2024] [Indexed: 02/04/2025]
Abstract
Chronic pain, as a social public health problem, has a serious impact on the quality of patients' life. Currently, the main drugs used to treat chronic pain are opioids, antipyretic, and nonsteroidal anti-inflammatory drugs (NSAIDs). But the obvious side effects limit their use, so it is urgent to find new therapeutic targets. Postsynaptic density (PSD)-95 protein plays an important role in the occurrence and development of chronic pain. The over-expression of the PSD-95 protein and its interaction with other proteins are closely related to the chronic pain. Besides, the PSD-95-related drugs that inhibit the expression of PSD-95 as well as the interaction with other protein have been proved to treat chronic pain significantly. In conclusion, although more deep studies are needed in the future, these studies indicate that PSD-95 and the related proteins, such as NMDA receptor (NMDAR) subunit 2B (GluN2B), AMPA receptor (AMPAR), calmodulin-dependent protein kinase II (CaMKII), 5-hydroxytryptamine 2A receptor (5-HT2AR), and neuronal nitric oxide synthase (nNOS), are the promising therapeutic targets for chronic pain.
Collapse
Affiliation(s)
- Lulin Ma
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Dongdong Sun
- Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Song Wen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jie Yuan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jing Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Xinran Tan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Song Cao
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China.
| |
Collapse
|
2
|
Brown CR, Shetty M, Foster JD. Palmitoylation regulates norepinephrine transporter uptake, surface localization, and total expression with pathogenic implications in postural orthostatic tachycardia syndrome. J Neurochem 2025; 169:e16241. [PMID: 39395208 PMCID: PMC11808474 DOI: 10.1111/jnc.16241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/14/2024]
Abstract
Postural orthostatic tachycardia syndrome (POTS) is an adrenergic signaling disorder characterized by excessive plasma norepinephrine, postural tachycardia, and syncope. The norepinephrine transporter (NET) modulates adrenergic homeostasis via the reuptake of extracellular catecholamines and is implicated in the pathogenesis of adrenergic and neurological disorders. In this study, we reveal NET is palmitoylated in male Sprague-Dawley rats and Lilly Laboratory Cell Porcine Kidney (LLC-PK1) cells. S-palmitoylation, or the addition of a 16-carbon saturated fatty acid, is a reversible post-translational modification responsible for the regulation of numerous biological mechanisms. We found that LLC-PK1 NET is dynamically palmitoylated, and that inhibition with the palmitoyl acyltransferase (DHHC) inhibitor, 2-bromopalmitate (2BP) results in decreased NET palmitoylation within 90 min of treatment. This result was followed closely by a reduction in transport capacity, cell surface, and total cellular NET expression after 120 min of treatment. Increasing 2BP concentrations and treatment time revealed a nearly complete loss of total NET protein. Co-expression with individual DHHCs revealed a single DHHC enzyme, DHHC1, promoted wild-type (WT) hNET palmitoylation and elevated NET protein levels. The POTS-associated NET mutant, A457P, exhibits dramatically decreased transport capacity and cell surface levels which we have confirmed in the current study. In an attempt to recover A457P NET expression, we co-expressed the A457P variant with DHHC1 to drive expression as seen with the WT protein but instead saw an increase in NET N-terminal immuno-detectable forms and fragments. Elimination of a potential palmitoylation site at cysteine 44 in the N-terminal tail of hNET resulted in a low expression phenotype mimicking the A457P hNET variant. Further investigation of A457P NET palmitoylation and surface expression is necessary, but our preliminary novel findings reveal palmitoylation as a mechanism of NET regulation and suggest that dysregulation of this process may contribute to the pathogenesis of adrenergic disorders like POTS.
Collapse
Affiliation(s)
- Christopher R. Brown
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health SciencesGrand ForksNorth DakotaUSA
| | - Madhur Shetty
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health SciencesGrand ForksNorth DakotaUSA
| | - James D. Foster
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health SciencesGrand ForksNorth DakotaUSA
| |
Collapse
|
3
|
Guan X, Wang T, Gao Y, Zhai H, Jiang F, Hou Q, Yang X, Wu H, Li LF, Luo Y, Li S, Sun Y, Qiu HJ, Li Y. The CP123L protein of African swine fever virus is a membrane-associated, palmitoylated protein required for viral replication. J Virol 2025; 99:e0144524. [PMID: 39714165 PMCID: PMC11784412 DOI: 10.1128/jvi.01445-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/31/2024] [Indexed: 12/24/2024] Open
Abstract
African swine fever (ASF) is a highly contagious and often lethal disease caused by African swine fever virus (ASFV) in pigs. Protein palmitoylation is a prevalent posttranslational lipid modification that can modulate viral replication. In this study, we investigated the palmitoylation of ASFV proteins. The results revealed that the CP123L protein (pCP123L) of ASFV was palmitoylated at the cysteine residue at position 18 (C18). To further elucidate the functional significance of this posttranslational modification, abolishing palmitoylation through a cysteine-to-serine mutation at C18 (C18S) of pCP123L (pCP123L/C18S) or treatment with 2-bromopalmitate (2-BP), a palmitoylation inhibitor, led to altered cytomembrane localization and migration rate of pCP123L. Furthermore, depalmitoylation achieved through 2-BP treatment significantly suppressed ASFV replication and exerted a profound impact on virus budding. Remarkably, blocking pCP123L palmitoylation via the C18S mutation resulted in decreased replication of ASFV. Our study represents the first evidence for the presence of palmitoylation in ASFV proteins and underscores its crucial role in viral replication. IMPORTANCE African swine fever (ASF) poses a significant threat to the global pig industry. The causative agent of ASF is African swine fever virus (ASFV), which encodes more than 165 proteins. Protein palmitoylation, a common posttranslational lipid modification, can modulate viral infection. To date, the ASFV proteins that undergo palmitoylation and their impacts on viral replication remain elusive. In this study, the CP123L protein (pCP123L) of ASFV was identified as a palmitoylated protein, and the cysteine residue at position 18 of pCP123L is responsible for its palmitoylation. Notably, our findings demonstrate that palmitoylation plays significant roles in ASFV protein functions and facilitates viral replication.
Collapse
Affiliation(s)
- Xiangyu Guan
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yuxuan Gao
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Huanjie Zhai
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Fengwei Jiang
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Qinghe Hou
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Xiaoke Yang
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Hongxia Wu
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yuzi Luo
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yuan Sun
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yongfeng Li
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| |
Collapse
|
4
|
Hohoff C, Kerkenberg N, Zhang M, Palkowska W, Wachsmuth L, Peng M, Stiehl L, Schettler C, Zang JCS, Huge A, Ponimaskin E, Faber C, Baune BT, Zhang W. Deficiency of the palmitoyl acyltransferase ZDHHC7 modulates depression-like behaviour in female mice after a mild chronic stress paradigm. Transl Psychiatry 2025; 15:20. [PMID: 39856044 PMCID: PMC11759705 DOI: 10.1038/s41398-025-03240-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/12/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Chronic stress (CS) is a debilitating condition that negatively affects body and brain. In mice, CS effects range from changes in behaviour and brain microstructure down to the level of gene expression. These effects are partly mediated by sex and sex steroid hormones, which in turn are affected by the palmitoyl acyltransferase ZDHHC7. ZDHHC7 might modulate also the response to CS via palmitoylation of sex steroid hormone receptors and other proteins critical for neuronal structure and functions. Therefore, we aimed to investigate the role of ZDHHC7 in response to CS on different system levels in a mouse model of Zdhhc7-deficiency. Female and male Zdhhc7-knockout (KO) and -wildtype (WT) mice underwent a four-week-mild CS paradigm or non-stress control (C) condition. After C or CS, behaviours, hippocampal microstructures (via MRI-based diffusion tensor imaging) and brain gene expression profiles (via mRNA-seq transcriptomics) were investigated. Analyses focused on effects of genotype (KO vs. WT) or condition (C vs. CS) separately in both sexes. Our results revealed significant effects particularly in females. Female KOs displayed increased locomotion and reduced depression-like behaviour after CS (KO vs. WT, C vs. CS: pall < 0.05). Hippocampal fibres were reduced in female KOs after C (KO vs. WT: pall < 0.05) but in female WTs after CS (C vs. CS: pall < 0.05). Furthermore, female KOs showed increased cortistatin expression after CS (C vs. CS: mRNAseq and qPCR pall < 0.05). In sum, Zdhhc7-deficiency reduced depression-like behaviours, prevented hippocampal fibre reduction and upregulated cortistatin after CS. It seemed to be related to a sex-specific stress response and may reveal genetic factors of CS-resilience in female mice.
Collapse
Affiliation(s)
- Christa Hohoff
- Department of Psychiatry, University of Münster, 48149, Münster, Germany.
| | - Nicole Kerkenberg
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
- Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, 48149, Münster, Germany
| | - Mingyue Zhang
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
| | - Weronika Palkowska
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
| | - Lydia Wachsmuth
- Clinic of Radiology, University of Münster, 48149, Münster, Germany
| | - Maja Peng
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
| | - Lena Stiehl
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
| | | | - Johannes C S Zang
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Huge
- Core Facility Genomics, University of Münster, 48149, Münster, Germany
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, 30625, Hannover, Germany
| | - Cornelius Faber
- Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, 48149, Münster, Germany
- Clinic of Radiology, University of Münster, 48149, Münster, Germany
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
- Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, 48149, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, University of Melbourne, Parkville, VIC, 3010, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Weiqi Zhang
- Department of Psychiatry, University of Münster, 48149, Münster, Germany
- Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, 48149, Münster, Germany
| |
Collapse
|
5
|
Huang B, Wang H, Liu S, Hao M, Luo D, Zhou Y, Huang Y, Nian Y, Zhang L, Chu B, Yin C. Palmitoylation-dependent regulation of GPX4 suppresses ferroptosis. Nat Commun 2025; 16:867. [PMID: 39833225 PMCID: PMC11746948 DOI: 10.1038/s41467-025-56344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
S-palmitoylation is a reversible and widespread post-translational modification, but its role in the regulation of ferroptosis has been poorly understood. Here, we elucidate that GPX4, an essential regulator of ferroptosis, is reversibly palmitoylated on cysteine 66. The acyltransferase ZDHHC20 palmitoylates GPX4 and increases its protein stability. ZDHHC20 depletion or inhibition of protein palmitoylation by 2-BP sensitizes cancer cells to ferroptosis. Moreover, we identify APT2 as the depalmitoylase of GPX4. Genetic silencing or pharmacological inhibition of APT2 with ML349 increases GPX4 palmitoylation, thereby stabilizing the protein and conferring resistance to ferroptosis. Notably, disrupting GPX4 palmitoylation markedly potentiates ferroptosis in xenografted and orthotopically implanted tumor models, and inhibits tumor metastasis through blood vessels. In the chemically induced colorectal cancer model, knockout of APT2 significantly aggravates cancer progression. Furthermore, pharmacologically modulating GPX4 palmitoylation impacts liver ischemia-reperfusion injury. Overall, our findings uncover the intricate network regulating GPX4 palmitoylation, highlighting its pivotal role in modulating ferroptosis sensitivity.
Collapse
Affiliation(s)
- Bin Huang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, Guangdong, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Hui Wang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China
| | - Shuo Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meng Hao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Dan Luo
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Yi Zhou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Ying Huang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Yong Nian
- College of Pharmacy, Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lei Zhang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, Guangdong, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chengqian Yin
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Dixon CL, Martin NR, Niphakis MJ, Cravatt BF, Fairn GD. Attenuating ABHD17 isoforms augments the S-acylation and function of NOD2 and a subset of Crohn's disease-associated NOD2 variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.20.572362. [PMID: 38187608 PMCID: PMC10769251 DOI: 10.1101/2023.12.20.572362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
BACKGROUND AND AIMS NOD2 is an intracellular innate immune receptor that detects bacterial peptidoglycan fragments. Although nominally soluble, some NOD2 is associated with the plasma membrane and endosomal compartments for microbial surveillance. This membrane targeting is achieved through post-translational S-acylation of NOD2 by the protein acyltransferase ZDHHC5. Membrane attachment is necessary to initiate a signaling cascade in response to cytosolic peptidoglycan fragments. Ultimately, this signaling results in the production of antimicrobial peptides and pro-inflammatory cytokines. In most cases, S-acylation is a reversible post-translational modification with removal of the fatty acyl chain catalyzed by one of several acyl protein thioesterases. Deacylation of NOD2 by such an enzyme will displace it from the plasma membrane and endosomes, thus preventing signaling. METHODS To identify the enzymes responsible for NOD2 deacylation, we used engineered cell lines with RNA interference and small-molecule inhibitors. These approaches were combined with confocal microscopy, acyl-resin-assisted capture, immunoblotting, and cytokine multiplex assays. RESULTS We identified α/β-hydrolase domain-containing protein 17 isoforms (ABHD17A, ABHD17B, and ABHD17C) as the acyl protein thioesterases responsible for NOD2 deacylation. Inhibiting ABHD17 increased the plasma membrane localization of wild-type NOD2 and a subset of poorly acylated Crohn's disease-associated variants. This enhanced NOD2 activity, increasing NF-κB activation and pro-inflammatory cytokine production in epithelial cells. CONCLUSIONS These findings demonstrate that ABHD17 isoforms are negative regulators of NOD2. The results also suggest that targeting ABHD17 isoforms could restore functionality to specific Crohn's disease-associated NOD2 variants, offering a potential therapeutic strategy.
Collapse
Affiliation(s)
- Charneal L. Dixon
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Noah R. Martin
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | | | | | - Gregory D. Fairn
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
7
|
Adachi N, Hess DT, Ueyama T. A facile assay for zDHHC palmitoyl transferase activation elucidates effects of mutation and modification. J Lipid Res 2025; 66:100743. [PMID: 39800157 DOI: 10.1016/j.jlr.2025.100743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
At least 10% of proteins constituting the human proteome are subject to S-acylation by a long-chain fatty acid, thioesterified to a Cys thiol side chain. Fatty S-acylation (prototypically, S-palmitoylation) operates across eukaryotic phylogeny and cell type. S-palmitoylation is carried out in mammalian cells by a family of 23-24 dedicated zDHHC palmitoyl transferase enzymes, and mutation of zDHHCs is associated with a number of human pathophysiologies. Activation of the zDHHCs by auto-S-palmitoylation, the transthioesterification of the active site Cys by fatty acyl coenzyme A, is the necessary first step in zDHHC-mediated protein S-palmitoylation. Most prior in vitro assessments of zDHHC activation have utilized purified zDHHCs, a time- and effort-intensive approach, which removes zDHHCs from their native membrane environment. We describe here a facile assay for zDHHC activation in native membranes. We overexpressed hemagglutinin-tagged wild-type or mutant zDHHCs in cultured HEK293 cells and prepared a whole membrane fraction, which was incubated with fluorescent palmitoyl CoA (NBD-palmitoyl-CoA) followed by SDS-PAGE, fluorescence imaging, and Western blotting for hemagglutinin. We show by mutational analysis that, as assayed, zDHHC auto-S-palmitoylation by NBD-palmitoyl-CoA is limited to the active site Cys. Application of the assay revealed differential effects on zDHHC activation of posttranslational zDHHC modification and of zDHHC mutations associated with human disease, in particular cancer. Our assay provides a facile means of assessing zDHHC activation, and thus of differentiating the effects of zDHHC mutation and posttranslational modification on zDHHC activation versus secondary effects on zDHHC functionality including altered zDHHC interaction with substrate palmitoyl-proteins.
Collapse
Affiliation(s)
- Naoko Adachi
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan.
| | - Douglas T Hess
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan.
| |
Collapse
|
8
|
Ding W, Gu J, Xu W, Wu J, Huang Y, Zhang S, Lin S. The Biosynthesis and Applications of Protein Lipidation. Chem Rev 2024; 124:12176-12212. [PMID: 39441663 DOI: 10.1021/acs.chemrev.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Protein lipidation dramatically affects protein structure, localization, and trafficking via remodeling protein-membrane and protein-protein interactions through hydrophobic lipid moieties. Understanding the biosynthesis of lipidated proteins, whether natural ones or mimetics, is crucial for reconstructing, validating, and studying the molecular mechanisms and biological functions of protein lipidation. In this Perspective, we first provide an overview of the natural enzymatic biosynthetic pathways of protein lipidation in mammalian cells, focusing on the enzymatic machineries and their chemical linkages. We then discuss strategies to biosynthesize protein lipidation in mammalian cells by engineering modification machineries and substrates. Additionally, we explore site-specific protein lipidation biosynthesis in vitro via enzyme-mediated ligations and in vivo primarily through genetic code expansion strategies. We also discuss the use of small molecule tools to modulate the process of protein lipidation biosynthesis. Finally, we provide concluding remarks and discuss future directions for the biosynthesis and applications of protein lipidation.
Collapse
Affiliation(s)
- Wenlong Ding
- Life Sciences Institute, Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
- Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jiayu Gu
- Department of Medical Oncology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenyuan Xu
- Life Sciences Institute, Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
| | - Jing Wu
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiwen Huang
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuai Zhang
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Shixian Lin
- Life Sciences Institute, Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Shaoxing Institute, Zhejiang University, Shaoxing 321000, China
- Department of Medical Oncology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
9
|
Gao M, Skolnick J. Predicting protein interactions of the kinase Lck critical to T cell modulation. Structure 2024; 32:2168-2179.e2. [PMID: 39368461 PMCID: PMC11560573 DOI: 10.1016/j.str.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 10/07/2024]
Abstract
Protein-protein interactions (PPIs) play pivotal roles in directing T cell fate. One key player is the non-receptor tyrosine protein kinase Lck that helps to transduce T cell activation signals. Lck is mediated by other proteins via interactions that are inadequately understood. Here, we use the deep learning method AF2Complex to predict PPIs involving Lck, by screening it against ∼1,000 proteins implicated in immune responses, followed by extensive structural modeling for selected interactions. Remarkably, we describe how Lck may be specifically targeted by a palmitoyltransferase using a phosphotyrosine motif. We uncover "hotspot" interactions between Lck and the tyrosine phosphatase CD45, leading to a significant conformational shift of Lck for activation. Lastly, we present intriguing interactions between the phosphotyrosine-binding domain of Lck and the cytoplasmic tail of the immune checkpoint LAG3 and propose a molecular mechanism for its inhibitory role. Together, this multifaceted study provides valuable insights into T cell regulation and signaling.
Collapse
Affiliation(s)
- Mu Gao
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; AgnistaBio Inc, Palo Alto, CA 94301, USA.
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
10
|
Wlodarczyk J, Bhattacharyya R, Dore K, Ho GPH, Martin DDO, Mejias R, Hochrainer K. Altered Protein Palmitoylation as Disease Mechanism in Neurodegenerative Disorders. J Neurosci 2024; 44:e1225242024. [PMID: 39358031 PMCID: PMC11450541 DOI: 10.1523/jneurosci.1225-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 10/04/2024] Open
Abstract
Palmitoylation, a lipid-based posttranslational protein modification, plays a crucial role in regulating various aspects of neuronal function through altering protein membrane-targeting, stabilities, and protein-protein interaction profiles. Disruption of palmitoylation has recently garnered attention as disease mechanism in neurodegeneration. Many proteins implicated in neurodegenerative diseases and associated neuronal dysfunction, including but not limited to amyloid precursor protein, β-secretase (BACE1), postsynaptic density protein 95, Fyn, synaptotagmin-11, mutant huntingtin, and mutant superoxide dismutase 1, undergo palmitoylation, and recent evidence suggests that altered palmitoylation contributes to the pathological characteristics of these proteins and associated disruption of cellular processes. In addition, dysfunction of enzymes that catalyze palmitoylation and depalmitoylation has been connected to the development of neurological disorders. This review highlights some of the latest advances in our understanding of palmitoylation regulation in neurodegenerative diseases and explores potential therapeutic implications.
Collapse
Affiliation(s)
- Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Raja Bhattacharyya
- Genetics and Aging Research Unit, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Kim Dore
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla, California 92093
| | - Gary P H Ho
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Dale D O Martin
- Department of Biology, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Rebeca Mejias
- Department of Physiology, School of Biology, Universidad de Sevilla, Seville, 41012 Spain
- Instituto de Investigaciones Biomédicas de Sevilla, IBIS/Universidad de Sevilla/Hospital Universitario Virgen del Rocío/Junta de Andalucía/CSIC, Seville 41013, Spain
| | - Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
11
|
Chen W, Guo L, Wei W, Cai C, Wu G. Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation is essential for maintenance of mammary stem cell activity. Cell Rep 2024; 43:114762. [PMID: 39321020 DOI: 10.1016/j.celrep.2024.114762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
Adult mammary stem cells (aMaSCs) are vital to tissue expansion and remodeling during the process of postnatal mammary development. The protein C receptor (Procr) is one of the well-identified surface markers of multipotent aMaSCs. However, an understanding of the regulatory mechanisms governing Procr's protein stability remains incomplete. In this study, we identified Glycoprotein m6a (Gpm6a) as a critical protein for aMaSC activity modulation by using the Gpm6a knockout mouse model. Interestingly, we determined that Gpm6a depletion results in a reduction of Procr protein stability. Mechanistically, Gpm6a regulates Procr protein stability by mediating the formation of lipid rafts, a process requiring Zdhhc1 and Zdhhc2 to palmitate Gpm6a at Cys17,18 and Cys246 sites. Our findings highlight an important mechanism involving Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation for the regulation of Procr stability, aMaSC activity, and postnatal mammary development.
Collapse
Affiliation(s)
- Weizhen Chen
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Luyao Guo
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Wei Wei
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
12
|
Yucel BP, Henley JM, Wilkinson KA. Protocol for detecting palmitoylation of high-molecular-weight rat synaptic proteins via acyl-PEG labeling. STAR Protoc 2024; 5:103296. [PMID: 39243378 PMCID: PMC11408371 DOI: 10.1016/j.xpro.2024.103296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/24/2024] [Accepted: 08/15/2024] [Indexed: 09/09/2024] Open
Abstract
Here, we present an optimized acyl-PEGyl exchange gel shift (APEGS) assay to monitor palmitoylation of high-molecular-weight proteins from primary neuronal cultures. We describe steps for culturing cortical neurons from rat embryos and expressing proteins of interest. We then detail procedures for employing a fatty acyl exchange technique wherein hydroxylamine is used to cleave palmitic acid from the palmitoyl-thioester bond, exposing cysteine residues that are subsequently labeled with methoxy polyethylene glycol maleimide (mPEG-MAL-10k). For complete details on the use and execution of this protocol, please refer to Yucel et al.1.
Collapse
Affiliation(s)
- Busra Perihan Yucel
- Centre for Synaptic Plasticity, School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, BS8 1TD Bristol, UK.
| | - Jeremy Martin Henley
- Centre for Synaptic Plasticity, School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, BS8 1TD Bristol, UK.
| | - Kevin Anthony Wilkinson
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, University Walk, BS8 1TD Bristol, UK.
| |
Collapse
|
13
|
Williams DM, Peden AA. S-acylation of NLRP3 provides a nigericin sensitive gating mechanism that controls access to the Golgi. eLife 2024; 13:RP94302. [PMID: 39263961 PMCID: PMC11392533 DOI: 10.7554/elife.94302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
NLRP3 is an inflammasome seeding pattern recognition receptor activated in response to multiple danger signals which perturb intracellular homeostasis. Electrostatic interactions between the NLRP3 polybasic (PB) region and negatively charged lipids on the trans-Golgi network (TGN) have been proposed to recruit NLRP3 to the TGN. In this study, we demonstrate that membrane association of NLRP3 is critically dependant on S-acylation of a highly conserved cysteine residue (Cys-130), which traps NLRP3 in a dynamic S-acylation cycle at the Golgi, and a series of hydrophobic residues preceding Cys-130 which act in conjunction with the PB region to facilitate Cys-130 dependent Golgi enrichment. Due to segregation from Golgi localised thioesterase enzymes caused by a nigericin induced breakdown in Golgi organisation and function, NLRP3 becomes immobilised on the Golgi through reduced de-acylation of its Cys-130 lipid anchor, suggesting that disruptions in Golgi homeostasis are conveyed to NLRP3 through its acylation state. Thus, our work defines a nigericin sensitive S-acylation cycle that gates access of NLRP3 to the Golgi.
Collapse
Affiliation(s)
- Daniel M Williams
- School of Bioscience, University of SheffieldSheffieldUnited Kingdom
| | - Andrew A Peden
- School of Bioscience, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
14
|
Koster KP, Green WN. Editorial: Role of protein palmitoylation in synaptic plasticity and neuronal differentiation, volume II. Front Synaptic Neurosci 2024; 16:1473989. [PMID: 39319198 PMCID: PMC11420002 DOI: 10.3389/fnsyn.2024.1473989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Affiliation(s)
- Kevin P. Koster
- Department of Neurobiology, University of Chicago, Chicago, IL, United States
| | | |
Collapse
|
15
|
P A H, Basavaraju N, Chandran M, Jaleel A, Bennett DA, Kommaddi RP. Mitigation of synaptic and memory impairments via F-actin stabilization in Alzheimer's disease. Alzheimers Res Ther 2024; 16:200. [PMID: 39244567 PMCID: PMC11380428 DOI: 10.1186/s13195-024-01558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/11/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Synaptic dysfunction, characterized by synapse loss and structural alterations, emerges as a prominent correlate of cognitive decline in Alzheimer's disease (AD). Actin cytoskeleton, which serves as the structural backbone of synaptic architecture, is observed to be lost from synapses in AD. Actin cytoskeleton loss compromises synaptic integrity, affecting glutamatergic receptor levels, neurotransmission, and synaptic strength. Understanding these molecular changes is crucial for developing interventions targeting synaptic dysfunction, potentially mitigating cognitive decline in AD. METHODS In this study, we investigated the synaptic actin interactome using mass spectrometry in a mouse model of AD, APP/PS1. Our objective was to explore how alterations in synaptic actin dynamics, particularly the interaction between PSD-95 and actin, contribute to synaptic and cognitive impairment in AD. To assess the impact of restoring F-actin levels on synaptic and cognitive functions in APP/PS1 mice, we administered F-actin stabilizing agent, jasplakinolide. Behavioral deficits in the mice were evaluated using the contextual fear conditioning paradigm. We utilized primary neuronal cultures to study the synaptic levels of AMPA and NMDA receptors and the dynamics of PSD-95 actin association. Furthermore, we analyzed postmortem brain tissue samples from subjects with no cognitive impairment (NCI), mild cognitive impairment (MCI), and Alzheimer's dementia (AD) to determine the association between PSD-95 and actin. RESULTS We found a significant reduction in PSD-95-actin association in synaptosomes from middle-aged APP/PS1 mice compared to wild-type (WT) mice. Treatment with jasplakinolide, an actin stabilizer, reversed deficits in memory recall, restored PSD-95-actin association, and increased synaptic F-actin levels in APP/PS1 mice. Additionally, actin stabilization led to elevated synaptic levels of AMPA and NMDA receptors, enhanced dendritic spine density, suggesting improved neurotransmission and synaptic strength in primary cortical neurons from APP/PS1 mice. Furthermore, analysis of postmortem human tissue with NCI, MCI and AD subjects revealed disrupted PSD-95-actin interactions, underscoring the clinical relevance of our preclinical studies. CONCLUSION Our study elucidates disrupted PSD-95 actin interactions across different models, highlighting potential therapeutic targets for AD. Stabilizing F-actin restores synaptic integrity and ameliorates cognitive deficits in APP/PS1 mice, suggesting that targeting synaptic actin regulation could be a promising therapeutic strategy to mitigate cognitive decline in AD.
Collapse
Affiliation(s)
- Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, Karnataka, 560012, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nimisha Basavaraju
- Centre for Brain Research, Indian Institute of Science, Bangalore, Karnataka, 560012, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - Mahesh Chandran
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Abdul Jaleel
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Reddy Peera Kommaddi
- Centre for Brain Research, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
| |
Collapse
|
16
|
Peng J, Liang D, Zhang Z. Palmitoylation of synaptic proteins: roles in functional regulation and pathogenesis of neurodegenerative diseases. Cell Mol Biol Lett 2024; 29:108. [PMID: 39127627 DOI: 10.1186/s11658-024-00625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Palmitoylation is a type of lipid modification that plays an important role in various aspects of neuronal function. Over the past few decades, several studies have shown that the palmitoylation of synaptic proteins is involved in neurotransmission and synaptic functions. Palmitoyl acyltransferases (PATs), which belong to the DHHC family, are major players in the regulation of palmitoylation. Dysregulated palmitoylation of synaptic proteins and mutated/dysregulated DHHC proteins are associated with several neurodegenerative diseases, such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD). In this review, we summarize the recent discoveries on the subcellular distribution of DHHC proteins and analyze their expression patterns in different brain cells. In particular, this review discusses how palmitoylation of synaptic proteins regulates synaptic vesicle exocytotic fusion and the localization, clustering, and transport of several postsynaptic receptors, as well as the role of palmitoylation of other proteins in regulating synaptic proteins. Additionally, some of the specific known associations of these factors with neurodegenerative disorders are explored, with a few suggestions for the development of therapeutic strategies. Finally, this review provides possible directions for future research to reveal detailed and specific mechanisms underlying the roles of synaptic protein palmitoylation.
Collapse
Affiliation(s)
- Jiaying Peng
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Danchan Liang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| |
Collapse
|
17
|
Liu Z, Li S, Wang C, Vidmar KJ, Bracey S, Li L, Willard B, Miyagi M, Lan T, Dickinson BC, Osme A, Pizarro TT, Xiao TS. Palmitoylation at a conserved cysteine residue facilitates gasdermin D-mediated pyroptosis and cytokine release. Proc Natl Acad Sci U S A 2024; 121:e2400883121. [PMID: 38980908 PMCID: PMC11260154 DOI: 10.1073/pnas.2400883121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Gasdermin D (GSDMD)-mediated pyroptotic cell death drives inflammatory cytokine release and downstream immune responses upon inflammasome activation, which play important roles in host defense and inflammatory disorders. Upon activation by proteases, the GSDMD N-terminal domain (NTD) undergoes oligomerization and membrane translocation in the presence of lipids to assemble pores. Despite intensive studies, the molecular events underlying the transition of GSDMD from an autoinhibited soluble form to an oligomeric pore form inserted into the membrane remain incompletely understood. Previous work characterized S-palmitoylation for gasdermins from bacteria, fungi, invertebrates, as well as mammalian gasdermin E (GSDME). Here, we report that a conserved residue Cys191 in human GSDMD was S-palmitoylated, which promoted GSDMD-mediated pyroptosis and cytokine release. Mutation of Cys191 or treatment with palmitoyltransferase inhibitors cyano-myracrylamide (CMA) or 2-bromopalmitate (2BP) suppressed GSDMD palmitoylation, its localization to the membrane and dampened pyroptosis or IL-1β secretion. Furthermore, Gsdmd-dependent inflammatory responses were alleviated by inhibition of palmitoylation in vivo. By contrast, coexpression of GSDMD with palmitoyltransferases enhanced pyroptotic cell death, while introduction of exogenous palmitoylation sequences fully restored pyroptotic activities to the C191A mutant, suggesting that palmitoylation-mediated membrane localization may be distinct from other molecular events such as GSDMD conformational change during pore assembly. Collectively, our study suggests that S-palmitoylation may be a shared regulatory mechanism for GSDMD and other gasdermins, which points to potential avenues for therapeutically targeting S-palmitoylation of gasdermins in inflammatory disorders.
Collapse
Affiliation(s)
- Zhonghua Liu
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Sai Li
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Chuanping Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Kaylynn J. Vidmar
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Syrena Bracey
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Ling Li
- Proteomics and Metabolic Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44196
| | - Belinda Willard
- Proteomics and Metabolic Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44196
| | - Masaru Miyagi
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH44106
| | - Tong Lan
- Department of Chemistry, University of Chicago, Chicago, IL60637
| | | | - Abdullah Osme
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Tsan Sam Xiao
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| |
Collapse
|
18
|
Voytyuk O, Ohata Y, Moustakas A, Ten Dijke P, Heldin CH. Smad7 palmitoylation by the S-acyltransferase zDHHC17 enhances its inhibitory effect on TGF-β/Smad signaling. J Biol Chem 2024; 300:107462. [PMID: 38876303 PMCID: PMC11277750 DOI: 10.1016/j.jbc.2024.107462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
Intracellular signaling by the pleiotropic cytokine transforming growth factor-β (TGF-β) is inhibited by Smad7 in a feedback control mechanism. The activity of Smad7 is tightly regulated by multiple post-translational modifications. Using resin-assisted capture and metabolic labeling methods, we show here that Smad7 is S-palmitoylated in mammary epithelial cell models that are widely studied because of their strong responses to TGF-β and their biological relevance to mammary development and tumor progression. S-palmitoylation of Smad7 is mediated by zDHHC17, a member of a family of 23 S-acyltransferase enzymes. Moreover, we identified four cysteine residues (Cys202, Cys225, Cys415, and Cys417) in Smad7 as palmitoylation acceptor sites. S-palmitoylation of Smad7 on Cys415 and Cys417 promoted the translocation of Smad7 from the nucleus to the cytoplasm, enhanced the stability of the Smad7 protein, and enforced its inhibitory effect on TGF-β-induced Smad transcriptional response. Thus, our findings reveal a new post-translational modification of Smad7, and highlight an important role of S-palmitoylation to enhance inhibition of TGF-β/Smad signaling by Smad7.
Collapse
Affiliation(s)
- Oleksandr Voytyuk
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, Uppsala, Sweden.
| | - Yae Ohata
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
S Mesquita F, Abrami L, Linder ME, Bamji SX, Dickinson BC, van der Goot FG. Mechanisms and functions of protein S-acylation. Nat Rev Mol Cell Biol 2024; 25:488-509. [PMID: 38355760 DOI: 10.1038/s41580-024-00700-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
Over the past two decades, protein S-acylation (often referred to as S-palmitoylation) has emerged as an important regulator of vital signalling pathways. S-Acylation is a reversible post-translational modification that involves the attachment of a fatty acid to a protein. Maintenance of the equilibrium between protein S-acylation and deacylation has demonstrated profound effects on various cellular processes, including innate immunity, inflammation, glucose metabolism and fat metabolism, as well as on brain and heart function. This Review provides an overview of current understanding of S-acylation and deacylation enzymes, their spatiotemporal regulation by sophisticated multilayered mechanisms, and their influence on protein function, cellular processes and physiological pathways. Furthermore, we examine how disruptions in protein S-acylation are associated with a broad spectrum of diseases from cancer to autoinflammatory disorders and neurological conditions.
Collapse
Affiliation(s)
- Francisco S Mesquita
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Laurence Abrami
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maurine E Linder
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | - Shernaz X Bamji
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - F Gisou van der Goot
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
20
|
Koster KP, Fyke Z, Nguyen TTA, Niqula A, Noriega-González LY, Woolfrey KM, Dell’Acqua ML, Cologna SM, Yoshii A. Akap5 links synaptic dysfunction to neuroinflammatory signaling in a mouse model of infantile neuronal ceroid lipofuscinosis. Front Synaptic Neurosci 2024; 16:1384625. [PMID: 38798824 PMCID: PMC11116793 DOI: 10.3389/fnsyn.2024.1384625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Palmitoylation and depalmitoylation represent dichotomic processes by which a labile posttranslational lipid modification regulates protein trafficking and degradation. The depalmitoylating enzyme, palmitoyl-protein thioesterase 1 (PPT1), is associated with the devastating pediatric neurodegenerative condition, infantile neuronal ceroid lipofuscinosis (CLN1). CLN1 is characterized by the accumulation of autofluorescent lysosomal storage material (AFSM) in neurons and robust neuroinflammation. Converging lines of evidence suggest that in addition to cellular waste accumulation, the symptomology of CLN1 corresponds with disruption of synaptic processes. Indeed, loss of Ppt1 function in cortical neurons dysregulates the synaptic incorporation of the GluA1 AMPA receptor (AMPAR) subunit during a type of synaptic plasticity called synaptic scaling. However, the mechanisms causing this aberration are unknown. Here, we used the Ppt1-/- mouse model (both sexes) to further investigate how Ppt1 regulates synaptic plasticity and how its disruption affects downstream signaling pathways. To this end, we performed a palmitoyl-proteomic screen, which provoked the discovery that Akap5 is excessively palmitoylated at Ppt1-/- synapses. Extending our previous data, in vivo induction of synaptic scaling, which is regulated by Akap5, caused an excessive upregulation of GluA1 in Ppt1-/- mice. This synaptic change was associated with exacerbated disease pathology. Furthermore, the Akap5- and inflammation-associated transcriptional regulator, nuclear factor of activated T cells (NFAT), was sensitized in Ppt1-/- cortical neurons. Suppressing the upstream regulator of NFAT activation, calcineurin, with the FDA-approved therapeutic FK506 (Tacrolimus) modestly improved neuroinflammation in Ppt1-/- mice. These findings indicate that the absence of depalmitoylation stifles synaptic protein trafficking and contributes to neuroinflammation via an Akap5-associated mechanism.
Collapse
Affiliation(s)
- Kevin P. Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zach Fyke
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Thu T. A. Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Amanda Niqula
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Kevin M. Woolfrey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Stephanie M. Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, United States
- Department of Neurology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
21
|
Brown CR, Foster JD. Palmitoylation regulates norepinephrine transporter trafficking and expression and is potentially involved in the pathogenesis of postural orthostatic tachycardia syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586171. [PMID: 38585862 PMCID: PMC10996475 DOI: 10.1101/2024.03.22.586171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Postural orthostatic tachycardia syndrome (POTS) is an adrenergic signaling disorder characterized by excessive plasma norepinephrine, postural tachycardia, and syncope. The norepinephrine transporter (NET) modulates adrenergic homeostasis via reuptake of extracellular catecholamines and is implicated in the pathogenesis of adrenergic and neurological disorders. Previous research has outlined that NET activity and trafficking is modulated via reversible post-translational modifications like phosphorylation and ubiquitylation. S-palmitoylation, or the addition of a 16-carbon saturated fatty acid, is another post-translational modification responsible for numerous biological mechanisms. In this study, we reveal that NET is dynamically palmitoylated and inhibition of this modification with the palmitoyl acyltransferase (DHHC) inhibitor, 2-bromopalmitate (2BP), results in decreased NET palmitoylation within 90 min of treatment. This result was followed closely with a reduction in transport capacity, cell surface, and total cellular NET expression after 120 min of treatment. Increasing 2BP concentrations and treatment time revealed a nearly complete loss of total NET protein. Co-expression with individual DHHCs revealed a single DHHC enzyme, DHHC1, promoted WT hNET palmitoylation and elevated NET protein levels. The POTS associated NET mutant, A457P, exhibits dramatically decreased transport capacity and cell surface levels which we have confirmed in the current study. In an attempt to recover A457P NET expression we co-expressed the A457P variant with DHHC1 to drive expression as seen with the WT protein but instead saw an increase in NET N-terminal immuno-detectable fragments. Further investigation of A457P NET palmitoylation and surface expression is necessary, but our preliminary novel findings reveal palmitoylation as a mechanism of NET regulation and suggest that dysregulation of this process may contribute to the pathogenesis of POTS.
Collapse
|
22
|
Binoy A, Kothari M, Sahadevan R, Poddar S, Kar P, Sadhukhan S. Protein S-palmitoylation is markedly inhibited by 4″-alkyl ether lipophilic derivatives of EGCG, the major green tea polyphenol: In vitro and in silico studies. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184264. [PMID: 38104647 DOI: 10.1016/j.bbamem.2023.184264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/27/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
S-palmitoylation is a dynamic lipid-based protein post-translational modification facilitated by a family of protein acyltransferases (PATs) commonly known as DHHC-PATs or DHHCs. It is the only lipid modification that is reversible, and this very fact uniquely qualifies it for therapeutic interventions through the development of DHHC inhibitors. Herein, we report that 4″-alkyl ether lipophilic derivatives of EGCG can effectively inhibit protein S-palmitoylation in vitro. With the help of metabolic labeling followed by copper(I)-catalyzed azide-alkyne cycloaddition Click reaction, we demonstrate that 4″-C14 EGCG and 4″-C16 EGCG markedly inhibited S-palmitoylation in various mammalian cells including HEK 293T, HeLa, and MCF-7 using both in gel fluorescence as well as confocal microscopy. Further, these EGCG derivatives were able to attenuate the S-palmitoylation to the basal level in DHHC3-overexpressed cells, suggesting that they are plausibly targeting DHHCs. Confocal microscopy data qualitatively reflected spatial and temporal distribution of S-palmitoylated proteins in different sub-cellular compartments and the inhibitory effects of 4″-C14 EGCG and 4″-C16 EGCG were clearly observed in the native cellular environment. Our findings were further substantiated by in silico analysis which revealed promising binding affinity and interactions of 4″-C14 EGCG and 4″-C16 EGCG with key amino acid residues present in the hydrophobic cleft of the DHHC20 enzyme. We also demonstrated the successful inhibition of S-palmitoylation of GAPDH by 4″-C16 EGCG. Taken together, our in vitro and in silico data strongly suggest that 4″-C14 EGCG and 4″-C16 EGCG can act as potent inhibitors for S-palmitoylation and can be employed as a complementary tool to investigate S-palmitoylation.
Collapse
Affiliation(s)
- Anupama Binoy
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala 678623, India
| | - Manan Kothari
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala 678623, India
| | - Revathy Sahadevan
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala 678623, India
| | - Sayan Poddar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Madhya Pradesh 453552, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Madhya Pradesh 453552, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Kerala 678623, India; Physical & Chemical Biology Laboratory, Indian Institute of Technology Palakkad, Kerala 678623, India; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Kerala 678623, India.
| |
Collapse
|
23
|
Essandoh K, Teuber JP, Brody MJ. Regulation of cardiomyocyte intracellular trafficking and signal transduction by protein palmitoylation. Biochem Soc Trans 2024; 52:41-53. [PMID: 38385554 PMCID: PMC10903464 DOI: 10.1042/bst20221296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/23/2024]
Abstract
Despite the well-established functions of protein palmitoylation in fundamental cellular processes, the roles of this reversible post-translational lipid modification in cardiomyocyte biology remain poorly studied. Palmitoylation is catalyzed by a family of 23 zinc finger and Asp-His-His-Cys domain-containing S-acyltransferases (zDHHC enzymes) and removed by select thioesterases of the lysophospholipase and α/β-hydroxylase domain (ABHD)-containing families of serine hydrolases. Recently, studies utilizing genetic manipulation of zDHHC enzymes in cardiomyocytes have begun to unveil essential functions for these enzymes in regulating cardiac development, homeostasis, and pathogenesis. Palmitoylation co-ordinates cardiac electrophysiology through direct modulation of ion channels and transporters to impact their trafficking or gating properties as well as indirectly through modification of regulators of channels, transporters, and calcium handling machinery. Not surprisingly, palmitoylation has roles in orchestrating the intracellular trafficking of proteins in cardiomyocytes, but also dynamically fine-tunes cardiomyocyte exocytosis and natriuretic peptide secretion. Palmitoylation has emerged as a potent regulator of intracellular signaling in cardiomyocytes, with recent studies uncovering palmitoylation-dependent regulation of small GTPases through direct modification and sarcolemmal targeting of the small GTPases themselves or by modification of regulators of the GTPase cycle. In addition to dynamic control of G protein signaling, cytosolic DNA is sensed and transduced into an inflammatory transcriptional output through palmitoylation-dependent activation of the cGAS-STING pathway, which has been targeted pharmacologically in preclinical models of heart disease. Further research is needed to fully understand the complex regulatory mechanisms governed by protein palmitoylation in cardiomyocytes and potential emerging therapeutic targets.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
| | - James P. Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Matthew J. Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, U.S.A
| |
Collapse
|
24
|
Kvergelidze E, Barbakadze T, Bátor J, Kalandadze I, Mikeladze D. Thyroid hormone T3 induces Fyn modification and modulates palmitoyltransferase gene expression through αvβ3 integrin receptor in PC12 cells during hypoxia. Transl Neurosci 2024; 15:20220347. [PMID: 39118829 PMCID: PMC11306964 DOI: 10.1515/tnsci-2022-0347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Thyroid hormones (THs) are essential in neuronal and glial cell development and differentiation, synaptogenesis, and myelin sheath formation. In addition to nuclear receptors, TH acts through αvβ3-integrin on the plasma membrane, influencing transcriptional regulation of signaling proteins that, in turn, affect adhesion and survival of nerve cells in various neurologic disorders. TH exhibits protective properties during brain hypoxia; however, precise intracellular mechanisms responsible for the preventive effects of TH remain unclear. In this study, we investigated the impact of TH on integrin αvβ3-dependent downstream systems in normoxic and hypoxic conditions of pheochromocytoma PC12 cells. Our findings reveal that triiodothyronine (T3), acting through αvβ3-integrin, induces activation of the JAK2/STAT5 pathway and suppression of the SHP2 in hypoxic PC12 cells. This activation correlates with the downregulation of the expression palmitoyltransferase-ZDHHC2 and ZDHHC9 genes, leading to a subsequent decrease in palmitoylation and phosphorylation of Fyn tyrosine kinase. We propose that these changes may occur due to STAT5-dependent epigenetic silencing of the palmitoyltransferase gene, which in turn reduces palmitoylation/phosphorylation of Fyn with a subsequent increase in the survival of cells. In summary, our study provides the first evidence demonstrating the involvement of integrin-dependent JAK/STAT pathway, SHP2 suppression, and altered post-translational modification of Fyn in protective effects of T3 during hypoxia.
Collapse
Affiliation(s)
- Elisabed Kvergelidze
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, 0162, Georgia
| | - Tamar Barbakadze
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, 0162, Georgia
- Laboratory of Biochemistry, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, 0160, Georgia
| | - Judit Bátor
- Department of Medical Biology and Central Electron Microscopic Laboratory, Medical School, University of Pécs, Pécs, 7624, Hungary
- Janos Szentagothai Research Centre, University of Pécs, Pécs, 7624, Hungary
| | - Irine Kalandadze
- Laboratory of Biochemistry, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, 0160, Georgia
| | - David Mikeladze
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, 0162, Georgia
- Laboratory of Biochemistry, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, 0160, Georgia
| |
Collapse
|
25
|
Traczyk G, Hromada-Judycka A, Świątkowska A, Wiśniewska J, Ciesielska A, Kwiatkowska K. Diacylglycerol kinase-ε is S-palmitoylated on cysteine in the cytoplasmic end of its N-terminal transmembrane fragment. J Lipid Res 2024; 65:100480. [PMID: 38008259 PMCID: PMC10759177 DOI: 10.1016/j.jlr.2023.100480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023] Open
Abstract
Diacylglycerol kinase-ε (DGKε) catalyzes phosphorylation of diacylglycerol to phosphatidic acid with a unique specificity toward 1-stearoyl-2-arachidonoyl-sn-glycerol, which is a backbone of phosphatidylinositol (PI). Owing to this specificity, DGKε is involved in the PI cycle maintaining the cellular level of phosphorylated PI derivatives of signaling activity and was also found crucial for lipid metabolism. DGKε dysfunction is linked with the development of atypical hemolytic uremic syndrome (aHUS) and possibly other human diseases. Despite the DGKε significance, data on its regulation by cotranslational and/or post-translational modifications are scarce. Here, we report that DGKε is S-palmitoylated at Cys38/40 (mouse/human DGKε) located in the cytoplasmic end of its N-terminal putative transmembrane fragment. The S-palmitoylation of DGKε was revealed by metabolic labeling of cells with a palmitic acid analogue followed by click chemistry and with acyl-biotin and acyl-polyethylene glycol exchange assays. The S-acyltransferases zDHHC7 (zinc finger DHHC domain containing) and zDHHC17 and the zDHHC6/16 tandem were found to catalyze DGKε S-palmitoylation, which also increased the DGKε abundance. Mouse DGKε-Myc ectopically expressed in human embryonic kidney 293 cells localized to the endoplasmic reticulum where zDHHC6/16 reside and in small amounts also to the Golgi apparatus where zDHHC7 and zDHHC17 are present. The Cys38Ala substitution upregulated, whereas hyperpalmitoylation of wild-type DGKε reduced the kinase activity, indicating an inhibitory effect of the Cys38 S-palmitoylation. In addition, the substitution of neighboring Pro31 with Ala also diminished the activity of DGKε. Taken together, our data indicate that S-palmitoylation can fine-tune DGKε activity in distinct cellular compartments, possibly by affecting the distance between the kinase and its substrate in a membrane.
Collapse
Affiliation(s)
- Gabriela Traczyk
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Aneta Hromada-Judycka
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Anna Świątkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Julia Wiśniewska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland.
| |
Collapse
|
26
|
Koster KP, Flores-Barrera E, Artur de la Villarmois E, Caballero A, Tseng KY, Yoshii A. Loss of Depalmitoylation Disrupts Homeostatic Plasticity of AMPARs in a Mouse Model of Infantile Neuronal Ceroid Lipofuscinosis. J Neurosci 2023; 43:8317-8335. [PMID: 37884348 PMCID: PMC10711723 DOI: 10.1523/jneurosci.1113-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Protein palmitoylation is the only reversible post-translational lipid modification. Palmitoylation is held in delicate balance by depalmitoylation to precisely regulate protein turnover. While over 20 palmitoylation enzymes are known, depalmitoylation is conducted by fewer enzymes. Of particular interest is the lack of the depalmitoylating enzyme palmitoyl-protein thioesterase 1 (PPT1) that causes the devastating pediatric neurodegenerative condition infantile neuronal ceroid lipofuscinosis (CLN1). While most of the research on Ppt1 function has centered on its role in the lysosome, recent findings demonstrated that many Ppt1 substrates are synaptic proteins, including the AMPA receptor (AMPAR) subunit GluA1. Still, the impact of Ppt1-mediated depalmitoylation on synaptic transmission and plasticity remains elusive. Thus, the goal of the present study was to use the Ppt1 -/- mouse model (both sexes) to determine whether Ppt1 regulates AMPAR-mediated synaptic transmission and plasticity, which are crucial for the maintenance of homeostatic adaptations in cortical circuits. Here, we found that basal excitatory transmission in the Ppt1 -/- visual cortex is developmentally regulated and that chemogenetic silencing of the Ppt1 -/- visual cortex excessively enhanced the synaptic expression of GluA1. Furthermore, triggering homeostatic plasticity in Ppt1 -/- primary neurons caused an exaggerated incorporation of GluA1-containing, calcium-permeable AMPARs, which correlated with increased GluA1 palmitoylation. Finally, Ca2+ imaging in awake Ppt1 -/- mice showed visual cortical neurons favor a state of synchronous firing. Collectively, our results elucidate a crucial role for Ppt1 in AMPAR trafficking and show that impeded proteostasis of palmitoylated synaptic proteins drives maladaptive homeostatic plasticity and abnormal recruitment of cortical activity in CLN1.SIGNIFICANCE STATEMENT Neuronal communication is orchestrated by the movement of receptors to and from the synaptic membrane. Protein palmitoylation is the only reversible post-translational lipid modification, a process that must be balanced precisely by depalmitoylation. The significance of depalmitoylation is evidenced by the discovery that mutation of the depalmitoylating enzyme palmitoyl-protein thioesterase 1 (Ppt1) causes severe pediatric neurodegeneration. In this study, we found that the equilibrium provided by Ppt1-mediated depalmitoylation is critical for AMPA receptor (AMPAR)-mediated plasticity and associated homeostatic adaptations of synaptic transmission in cortical circuits. This finding complements the recent explosion of palmitoylation research by emphasizing the necessity of balanced depalmitoylation.
Collapse
Affiliation(s)
- Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Eden Flores-Barrera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | | | - Adriana Caballero
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois 60612
- Department of Neurology, University of Illinois at Chicago, Chicago, Illinois 60612
| |
Collapse
|
27
|
Liu H, Yan P, Wu C, Rao M, Zhu J, Lv L, Li W, Liang Y, Qi S, Lu K, Kong E. Palmitoylated Sept8-204 modulates learning and anxiety by regulating filopodia arborization and actin dynamics. Sci Signal 2023; 16:eadi8645. [PMID: 38051778 DOI: 10.1126/scisignal.adi8645] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023]
Abstract
Septin proteins are involved in diverse physiological functions, including the formation of specialized cytoskeletal structures. Septin 8 (Sept8) is implicated in spine morphogenesis and dendritic branching through palmitoylation. We explored the role and regulation of a Sept8 variant in human neural-like cells and in the mouse brain. We identified Sept8-204 as a brain-specific variant of Sept8 that was abundant in neurons and modified by palmitoylation, specifically at Cys469, Cys470, and Cys472. Sept8-204 palmitoylation was mediated by the palmitoyltransferase ZDHHC7 and was removed by the depalmitoylase PPT1. Palmitoylation of Sept8-204 bound to F-actin and induced cytoskeletal dynamics to promote the outgrowth of filopodia in N2a cells and the arborization of neurites in hippocampal neurons. In contrast, a Sept8-204 variant that could not be palmitoylated because of mutation of all three Cys residues (Sept8-204-3CA) lost its ability to bind F-actin, and expression of this mutant did not promote morphological changes. Genetic deletion of Sept8, Sept8-204, or Zdhhc7 caused deficits in learning and memory and promoted anxiety-like behaviors in mice. Our findings provide greater insight into the regulation of Sept8-204 by palmitoylation and its role in neuronal morphology and function in relation to cognition.
Collapse
Affiliation(s)
- Huicong Liu
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang 453003, China
| | - Peipei Yan
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang 453003, China
| | - Can Wu
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang 453003, China
| | - Muding Rao
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang 453003, China
| | - Jiangli Zhu
- Department of Urology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu 610041, China
| | - Luxian Lv
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Wenqiang Li
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yinming Liang
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang 453003, China
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu 610041, China
| | - Kefeng Lu
- Department of Neurology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Eryan Kong
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
28
|
Baldwin TA, Teuber JP, Kuwabara Y, Subramani A, Lin SCJ, Kanisicak O, Vagnozzi RJ, Zhang W, Brody MJ, Molkentin JD. Palmitoylation-dependent regulation of cardiomyocyte Rac1 signaling activity and minor effects on cardiac hypertrophy. J Biol Chem 2023; 299:105426. [PMID: 37926281 PMCID: PMC10716590 DOI: 10.1016/j.jbc.2023.105426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
S-palmitoylation is a reversible lipid modification catalyzed by 23 S-acyltransferases with a conserved zinc finger aspartate-histidine-histidine-cysteine (zDHHC) domain that facilitates targeting of proteins to specific intracellular membranes. Here we performed a gain-of-function screen in the mouse and identified the Golgi-localized enzymes zDHHC3 and zDHHC7 as regulators of cardiac hypertrophy. Cardiomyocyte-specific transgenic mice overexpressing zDHHC3 show cardiac disease, and S-acyl proteomics identified the small GTPase Rac1 as a novel substrate of zDHHC3. Notably, cardiomyopathy and congestive heart failure in zDHHC3 transgenic mice is preceded by enhanced Rac1 S-palmitoylation, membrane localization, activity, downstream hypertrophic signaling, and concomitant induction of all Rho family small GTPases whereas mice overexpressing an enzymatically dead zDHHC3 mutant show no discernible effect. However, loss of Rac1 or other identified zDHHC3 targets Gαq/11 or galectin-1 does not diminish zDHHC3-induced cardiomyopathy, suggesting multiple effectors and pathways promoting decompensation with sustained zDHHC3 activity. Genetic deletion of Zdhhc3 in combination with Zdhhc7 reduces cardiac hypertrophy during the early response to pressure overload stimulation but not over longer time periods. Indeed, cardiac hypertrophy in response to 2 weeks of angiotensin-II infusion is not diminished by Zdhhc3/7 deletion, again suggesting other S-acyltransferases or signaling mechanisms compensate to promote hypertrophic signaling. Taken together, these data indicate that the activity of zDHHC3 and zDHHC7 at the cardiomyocyte Golgi promote Rac1 signaling and maladaptive cardiac remodeling, but redundant signaling effectors compensate to maintain cardiac hypertrophy with sustained pathological stimulation in the absence of zDHHC3/7.
Collapse
Affiliation(s)
- Tanya A Baldwin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James P Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yasuhide Kuwabara
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Araskumar Subramani
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Suh-Chin J Lin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Onur Kanisicak
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ronald J Vagnozzi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Division of Cardiology, Department of Medicine, Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Mental Health, University of Münster, Münster, Germany
| | - Matthew J Brody
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
29
|
Meng X, Templeton C, Clementi C, Veit M. The role of an amphiphilic helix and transmembrane region in the efficient acylation of the M2 protein from influenza virus. Sci Rep 2023; 13:18928. [PMID: 37919373 PMCID: PMC10622425 DOI: 10.1038/s41598-023-45945-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Protein palmitoylation, a cellular process occurring at the membrane-cytosol interface, is orchestrated by members of the DHHC enzyme family and plays a pivotal role in regulating various cellular functions. The M2 protein of the influenza virus, which is acylated at a membrane-near amphiphilic helix serves as a model for studying the intricate signals governing acylation and its interaction with the cognate enzyme, DHHC20. We investigate it here using both experimental and computational assays. We report that altering the biophysical properties of the amphiphilic helix, particularly by shortening or disrupting it, results in a substantial reduction in M2 palmitoylation, but does not entirely abolish the process. Intriguingly, DHHC20 exhibits an augmented affinity for some M2 mutants compared to the wildtype M2. Molecular dynamics simulations unveil interactions between amino acids of the helix and the catalytically significant DHHC and TTXE motifs of DHHC20. Our findings suggest that the binding of M2 to DHHC20, while not highly specific, is mediated by requisite contacts, possibly instigating the transfer of fatty acids. A comprehensive comprehension of protein palmitoylation mechanisms is imperative for the development of DHHC-specific inhibitors, holding promise for the treatment of diverse human diseases.
Collapse
Affiliation(s)
- Xiaorong Meng
- Institute of Virology, Veterinary Faculty, Freie Universität Berlin, Berlin, Germany
| | - Clark Templeton
- Theoretical and Computational Biophysics, Department of Physics, Freie Universität Berlin, Berlin, Germany
| | - Cecilia Clementi
- Theoretical and Computational Biophysics, Department of Physics, Freie Universität Berlin, Berlin, Germany
| | - Michael Veit
- Institute of Virology, Veterinary Faculty, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
30
|
Meng X, Veit M. Palmitoylation of the hemagglutinin of influenza B virus by ER-localized DHHC enzymes 1, 2, 4, and 6 is required for efficient virus replication. J Virol 2023; 97:e0124523. [PMID: 37792001 PMCID: PMC10617437 DOI: 10.1128/jvi.01245-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Influenza viruses are a public health concern since they cause seasonal outbreaks and occasionally pandemics. Our study investigates the importance of a protein modification called "palmitoylation" in the replication of influenza B virus. Palmitoylation involves attaching fatty acids to the viral protein hemagglutinin and has previously been studied for influenza A virus. We found that this modification is important for the influenza B virus to replicate, as mutating the sites where palmitate is attached prevented the virus from generating viable particles. Our experiments also showed that this modification occurs in the endoplasmic reticulum. We identified the specific enzymes responsible for this modification, which are different from those involved in palmitoylation of HA of influenza A virus. Overall, our research illuminates the similarities and differences in fatty acid attachment to HA of influenza A and B viruses and identifies the responsible enzymes, which might be promising targets for anti-viral therapy.
Collapse
Affiliation(s)
- Xiaorong Meng
- Veterinary Faculty, Institute for Virology, Freie Universität Berlin , Berlin, Germany
| | - Michael Veit
- Veterinary Faculty, Institute for Virology, Freie Universität Berlin , Berlin, Germany
| |
Collapse
|
31
|
Teo S, Bossio A, Stamatakou E, Pascual-Vargas P, Jones ME, Schuhmacher LN, Salinas PC. S-acylation of the Wnt receptor Frizzled-5 by zDHHC5 controls its cellular localization and synaptogenic activity in the rodent hippocampus. Dev Cell 2023; 58:2063-2079.e9. [PMID: 37557176 DOI: 10.1016/j.devcel.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 05/05/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
Proper localization of receptors for synaptic organizing factors is crucial for synapse formation. Wnt proteins promote synapse assembly through Frizzled (Fz) receptors. In hippocampal neurons, the surface and synaptic localization of Fz5 is regulated by neuronal activity, but the mechanisms involved remain poorly understood. Here, we report that all Fz receptors can be post-translationally modified by S-acylation and that Fz5 is S-acylated on three C-terminal cysteines by zDHHC5. S-acylation is essential for Fz5 localization to the cell surface, axons, and presynaptic sites. Notably, S-acylation-deficient Fz5 is internalized faster, affecting its association with signalosome components at the cell surface. S-acylation-deficient Fz5 also fails to activate canonical and divergent canonical Wnt pathways. Fz5 S-acylation levels are regulated by the pattern of neuronal activity. In vivo studies demonstrate that S-acylation-deficient Fz5 expression fails to induce presynaptic assembly. Our studies show that S-acylation of Frizzled receptors is a mechanism controlling their localization and function.
Collapse
Affiliation(s)
- Samuel Teo
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Alessandro Bossio
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Eleanna Stamatakou
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Patricia Pascual-Vargas
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Megan E Jones
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Laura-Nadine Schuhmacher
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Patricia C Salinas
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London WC1E 6BT, UK.
| |
Collapse
|
32
|
Wang G, Ji X, Nie L, Xu R. Exploring the proteins and metabolites associated with male antennae responses to female exposure of Antheraea pernyi (Lepidoptera: Saturniidae) moths. JOURNAL OF ECONOMIC ENTOMOLOGY 2023; 116:1838-1849. [PMID: 37459048 DOI: 10.1093/jee/toad132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/13/2023] [Accepted: 06/24/2023] [Indexed: 10/12/2023]
Abstract
Detection of sex pheromones of insects relies on the antennae. The female pheromone signal transmission in the male antennae ultimately initiates the courtship and mating behaviors of males. To investigate the proteins and metabolites involved in this neural transduction, integrative proteomics and metabolomics analysis including tandem mass tag (TMT) proteomic quantification and liquid chromatography tandem mass spectrometry (LC/MS)-based metabolomics was adopted for comparing proteomic and metabolic changes between the antennae of male moths following stimulation by females and the non-stimulated males of Antheraea pernyi (Guérin-Méneville, Lepidoptera: Saturniidae) in this study. A total of 92 differentially expressed proteins (DEPs) containing 52 upregulated and 40 downregulated proteins and 545 differentially expressed metabolites (DEMs) including 218 upregulated and 327 downregulated metabolites were identified from the antennae of female-stimulated male moths based on the proteome and metabolome data, respectively. Bioinformatics analysis was performed for the 45 DEPs and 160 DEMs, including Gene Ontology (GO), Clusters of Orthologous Groups (COG), and Kyoto Encylopaedia of Genes and Genomes (KEGG) enrichment analysis and Human Metabolome Database (HMDB) annotation. A number of DEPs and DEMs related to neural transmission of female pheromone signals in the male antennae of A. pernyi were screened, including tyrosine hydroxylase, cryptochrome-1, tachykinin, arylalkylamine N-acetyltransferase, cadherin-23, glutathione S-transferase delta 3, tyramine, tryptamine, n-oleoyl dopamine, n-stearoyl dopamine, and n-stearoyl tyrosine. The altered expression levels of those proteins or metabolites were speculated involved in regulating the neuron activity for enhanced transmission of neural impulses and continuous perception, reception, and transduction of female pheromone signals. Our findings yielded novel insights into the potential mechanisms in the antennae of male A. pernyi responding to female attraction.
Collapse
Affiliation(s)
- Guobao Wang
- College of Biology and Oceanography, Weifang University, Weifang 261061, China
| | - Xiang Ji
- College of Biology and Oceanography, Weifang University, Weifang 261061, China
| | - Lei Nie
- Shandong Sericulture Research Institute, Shandong Academy of Agricultural Sciences, Yantai 264002, China
| | - Ruirui Xu
- College of Biology and Oceanography, Weifang University, Weifang 261061, China
| |
Collapse
|
33
|
Salaun C, Tomkinson NCO, Chamberlain LH. The endoplasmic reticulum-localized enzyme zDHHC6 mediates S-acylation of short transmembrane constructs from multiple type I and II membrane proteins. J Biol Chem 2023; 299:105201. [PMID: 37660915 PMCID: PMC10520890 DOI: 10.1016/j.jbc.2023.105201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023] Open
Abstract
In this study, we investigated the S-acylation of two host cell proteins important for viral infection: TMPRSS2 (transmembrane serine protease 2), which cleaves severe acute respiratory syndrome coronavirus 2 spike to facilitate viral entry, and bone marrow stromal antigen 2, a general viral restriction factor. We found that both proteins were S-acylated by zDHHC6, an S-acyltransferase enzyme localized at the endoplasmic reticulum, in coexpression experiments. Mutagenic analysis revealed that zDHHC6 modifies a single cysteine in each protein, which are in proximity to the transmembrane domains (TMDs). For TMPRSS2, the modified cysteine is positioned two residues into the TMD, whereas the modified cysteine in bone marrow stromal antigen 2 has a cytosolic location two amino acids upstream of the TMD. Cysteine swapping revealed that repositioning the target cysteine of TMPRSS2 further into the TMD substantially reduced S-acylation by zDHHC6. Interestingly, zDHHC6 efficiently S-acylated truncated forms of these proteins that contained only the TMDs and short juxtamembrane regions. The ability of zDHHC6 to modify short TMD sequences was also seen for the transferrin receptor (another type II membrane protein) and for five different type I membrane protein constructs, including cluster of differentiation 4. Collectively, the results of this study show that zDHHC6 can modify diverse membrane proteins (type I and II) and requires only the presence of the TMD and target cysteine for efficient S-acylation. Thus, zDHHC6 may be a broad specificity S-acyltransferase specialized for the modification of a diverse set of transmembrane proteins at the endoplasmic reticulum.
Collapse
Affiliation(s)
- Christine Salaun
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom.
| | - Nicholas C O Tomkinson
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Luke H Chamberlain
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
34
|
Shetty M, Bolland DE, Morrell J, Grove BD, Foster JD, Vaughan RA. Dopamine transporter membrane mobility is bidirectionally regulated by phosphorylation and palmitoylation. Curr Res Physiol 2023; 6:100106. [PMID: 38107792 PMCID: PMC10724222 DOI: 10.1016/j.crphys.2023.100106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023] Open
Abstract
The primary regulator of dopamine availability in the brain is the dopamine transporter (DAT), a plasma membrane protein that drives reuptake of released dopamine from the extracellular space into the presynaptic neuron. DAT activity is regulated by post-translational modifications that establish clearance capacity through impacts on transport kinetics, and dysregulation of these events may underlie dopaminergic imbalances in mood and psychiatric disorders. Here, using fluorescence recovery after photobleaching, we show that phosphorylation and palmitoylation induce opposing effects on DAT lateral membrane mobility, which may influence functional outcomes by regulating subcellular localization and binding partner interactions. Membrane mobility was also impacted by amphetamine and in polymorphic variant A559V in directions consistent with enhanced phosphorylation. These findings grow the list of DAT properties controlled by these post-translational modifications and highlight their role in establishment of dopaminergic tone in physiological and pathophysiological states.
Collapse
Affiliation(s)
- Madhur Shetty
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | | | - Joshua Morrell
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Bryon D. Grove
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| |
Collapse
|
35
|
Brown CR, Foster JD. Palmitoylation Regulates Human Serotonin Transporter Activity, Trafficking, and Expression and Is Modulated by Escitalopram. ACS Chem Neurosci 2023; 14:3431-3443. [PMID: 37644775 DOI: 10.1021/acschemneuro.3c00319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
In the central nervous system, serotonergic signaling modulates sleep, mood, and cognitive control. During serotonergic transmission, the synaptic concentration of serotonin is tightly controlled in a spatial and temporal manner by the serotonin transporter (SERT). Dysregulation of this process is implicated in the pathogenesis of major-depressive, obsessive-compulsive, and autism-spectrum disorders, which makes SERT a primary target for prescription therapeutics, most notably selective serotonin reuptake inhibitors (SSRIs). S-Palmitoylation, the reversible addition of a 16-carbon fatty acid to proteins, is an increasingly recognized dynamic post-translational modification responsible for modulating protein kinetics, trafficking, and localization patterns in response to physiologic/cellular stimuli. In this study, we reveal that human SERTs are a target for palmitoylation, and using the irreversible palmitoyl acyltransferase inhibitor 2-bromopalmitate (2BP), we have identified several associated functions. Using a lower dose of 2BP in shorter time frames, inhibition of palmitoylation was associated with reductions in SERT Vmax, without changes in Km or surface expression. With higher doses of 2BP for longer time intervals, inhibition of palmitoylation was consistent with the loss of cell surface and total SERT protein, suggesting palmitoylation is an important mechanism in regulating SERT trafficking and maintenance of SERT protein through biogenic or anti-degradative processes. Additionally, we have identified that treatment with the SSRI escitalopram decreases SERT palmitoylation analogous to 2BP, reducing SERT surface expression and transport capacity. Ultimately, these results reveal that palmitoylation is a major regulatory mechanism for SERT kinetics and trafficking and may be the mechanism responsible for escitalopram-induced internalization and ultimately decreased cellular SERT protein levels.
Collapse
Affiliation(s)
- Christopher R Brown
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, North Dakota 58202, United States
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, North Dakota 58202, United States
| |
Collapse
|
36
|
Li MD, Wang L, Zheng YQ, Huang DH, Xia ZX, Liu JM, Tian D, OuYang H, Wang ZH, Huang Z, Lin XS, Zhu XQ, Wang SY, Chen WK, Yang SW, Zhao YL, Liu JA, Shen ZC. DHHC2 regulates fear memory formation, LTP, and AKAP150 signaling in the hippocampus. iScience 2023; 26:107561. [PMID: 37664599 PMCID: PMC10469764 DOI: 10.1016/j.isci.2023.107561] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/07/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Palmitoyl acyltransferases (PATs) have been suggested to be involved in learning and memory. However, the underlying mechanisms have not yet been fully elucidated. Here, we found that the activity of DHHC2 was upregulated in the hippocampus after fear conditioning, and DHHC2 knockdown impaired fear induced memory and long-term potentiation (LTP). Additionally, the activity of DHHC2 and its synaptic expression were increased after high frequency stimulation (HFS) or glycine treatment. Importantly, fear learning selectively augmented the palmitoylation level of AKAP150, not PSD-95, and this effect was abolished by DHHC2 knockdown. Furthermore, 2-bromopalmitic acid (2-BP), a palmitoylation inhibitor, attenuated the increased palmitoylation level of AKAP150 and the interaction between AKAP150 and PSD-95 induced by HFS. Lastly, DHHC2 knockdown reduced the phosphorylation level of GluA1 at Ser845, and also induced an impairment of LTP in the hippocampus. Our results suggest that DHHC2 plays a critical role in regulating fear memory via AKAP150 signaling.
Collapse
Affiliation(s)
- Meng-Die Li
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lu Wang
- Department of Nephrology, Fuzhou Children’s Hospital of Fujian Province, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Yu-Qi Zheng
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Dan-Hong Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zhi-Xuan Xia
- Department of Pharmacology, School of Basic Medicine and Life Science, Hainan Medical University, Haikou 571199, China
| | - Jian-Min Liu
- Department of Pharmacy, Wuhan No. 1 Hospital, Wuhan 430000, China
| | - Dan Tian
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Hui OuYang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zi-Hao Wang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhen Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Xiao-Shan Lin
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Xiao-Qian Zhu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Si-Ying Wang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Wei-Kai Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Shao-Wei Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yue-Ling Zhao
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jia-An Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zu-Cheng Shen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
37
|
Villanueva CE, Hagenbuch B. Palmitoylation of solute carriers. Biochem Pharmacol 2023; 215:115695. [PMID: 37481134 PMCID: PMC10530500 DOI: 10.1016/j.bcp.2023.115695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Post-translational modifications are an important mechanism in the regulation of protein expression, function, and degradation. Well-known post-translational modifications are phosphorylation, glycosylation, and ubiquitination. However, lipid modifications, including myristoylation, prenylation, and palmitoylation, are poorly studied. Since the early 2000s, researchers have become more interested in lipid modifications, especially palmitoylation. The number of articles in PubMed increased from about 350 between 2000 and 2005 to more than 600 annually during the past ten years. S-palmitoylation, where the 16-carbon saturated (C16:0) palmitic acid is added to free cysteine residues of proteins, is a reversible protein modification that can affect the expression, membrane localization, and function of the modified proteins. Various diseases like Huntington's and Alzheimer's disease have been linked to changes in protein palmitoylation. In humans, the addition of palmitic acid is mediated by 23 palmitoyl acyltransferases, also called DHHC proteins. The modification can be reversed by a few thioesterases or hydrolases. Numerous soluble and membrane-attached proteins are known to be palmitoylated, but among the approximately 400 solute carriers that are classified in 66 families, only 15 found in 8 families have so far been documented to be palmitoylated. Among the best-characterized transporters are the glucose transporters GLUT1 (SLC2A1) and GLUT4 (SLC2A4), the three monoamine transporters norepinephrine transporter (NET; SLC6A2), dopamine transporter (DAT; SLC6A3), and serotonin transporter (SERT; SLC6A4), and the sodium-calcium exchanger NCX1 (SLC8A1). While there is evidence from recent proteomics experiments that numerous solute carriers are palmitoylated, no details beyond the 15 transporters covered in this review are available.
Collapse
Affiliation(s)
- Cecilia E Villanueva
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
38
|
Li X, Gluth A, Zhang T, Qian WJ. Thiol redox proteomics: Characterization of thiol-based post-translational modifications. Proteomics 2023; 23:e2200194. [PMID: 37248656 PMCID: PMC10764013 DOI: 10.1002/pmic.202200194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
Redox post-translational modifications on cysteine thiols (redox PTMs) have profound effects on protein structure and function, thus enabling regulation of various biological processes. Redox proteomics approaches aim to characterize the landscape of redox PTMs at the systems level. These approaches facilitate studies of condition-specific, dynamic processes implicating redox PTMs and have furthered our understanding of redox signaling and regulation. Mass spectrometry (MS) is a powerful tool for such analyses which has been demonstrated by significant advances in redox proteomics during the last decade. A group of well-established approaches involves the initial blocking of free thiols followed by selective reduction of oxidized PTMs and subsequent enrichment for downstream detection. Alternatively, novel chemoselective probe-based approaches have been developed for various redox PTMs. Direct detection of redox PTMs without any enrichment has also been demonstrated given the sensitivity of contemporary MS instruments. This review discusses the general principles behind different analytical strategies and covers recent advances in redox proteomics. Several applications of redox proteomics are also highlighted to illustrate how large-scale redox proteomics data can lead to novel biological insights.
Collapse
Affiliation(s)
- Xiaolu Li
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354
| | - Austin Gluth
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354
- Department of Biological Systems Engineering, Washington State University, Richland, WA 99354
| | - Tong Zhang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354
| |
Collapse
|
39
|
Ren JG, Xing B, Lv K, O’Keefe RA, Wu M, Wang R, Bauer KM, Ghazaryan A, Burslem GM, Zhang J, O’Connell RM, Pillai V, Hexner EO, Philips MR, Tong W. RAB27B controls palmitoylation-dependent NRAS trafficking and signaling in myeloid leukemia. J Clin Invest 2023; 133:e165510. [PMID: 37317963 PMCID: PMC10266782 DOI: 10.1172/jci165510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/24/2023] [Indexed: 06/16/2023] Open
Abstract
RAS mutations are among the most prevalent oncogenic drivers in cancers. RAS proteins propagate signals only when associated with cellular membranes as a consequence of lipid modifications that impact their trafficking. Here, we discovered that RAB27B, a RAB family small GTPase, controlled NRAS palmitoylation and trafficking to the plasma membrane, a localization required for activation. Our proteomic studies revealed RAB27B upregulation in CBL- or JAK2-mutated myeloid malignancies, and its expression correlated with poor prognosis in acute myeloid leukemias (AMLs). RAB27B depletion inhibited the growth of CBL-deficient or NRAS-mutant cell lines. Strikingly, Rab27b deficiency in mice abrogated mutant but not WT NRAS-mediated progenitor cell growth, ERK signaling, and NRAS palmitoylation. Further, Rab27b deficiency significantly reduced myelomonocytic leukemia development in vivo. Mechanistically, RAB27B interacted with ZDHHC9, a palmitoyl acyltransferase that modifies NRAS. By regulating palmitoylation, RAB27B controlled c-RAF/MEK/ERK signaling and affected leukemia development. Importantly, RAB27B depletion in primary human AMLs inhibited oncogenic NRAS signaling and leukemic growth. We further revealed a significant correlation between RAB27B expression and sensitivity to MEK inhibitors in AMLs. Thus, our studies presented a link between RAB proteins and fundamental aspects of RAS posttranslational modification and trafficking, highlighting future therapeutic strategies for RAS-driven cancers.
Collapse
Affiliation(s)
- Jian-Gang Ren
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bowen Xing
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kaosheng Lv
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry, School of Medicine at the Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Rachel A. O’Keefe
- Department of Medicine and Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Mengfang Wu
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ruoxing Wang
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kaylyn M. Bauer
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Arevik Ghazaryan
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - George M. Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Ryan M. O’Connell
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Vinodh Pillai
- Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Elizabeth O. Hexner
- Division of Hematology and Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark R. Philips
- Department of Medicine and Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Wei Tong
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
40
|
Ramzan F, Abrar F, Mishra GG, Liao LMQ, Martin DDO. Lost in traffic: consequences of altered palmitoylation in neurodegeneration. Front Physiol 2023; 14:1166125. [PMID: 37324388 PMCID: PMC10268010 DOI: 10.3389/fphys.2023.1166125] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
One of the first molecular events in neurodegenerative diseases, regardless of etiology, is protein mislocalization. Protein mislocalization in neurons is often linked to proteostasis deficiencies leading to the build-up of misfolded proteins and/or organelles that contributes to cellular toxicity and cell death. By understanding how proteins mislocalize in neurons, we can develop novel therapeutics that target the earliest stages of neurodegeneration. A critical mechanism regulating protein localization and proteostasis in neurons is the protein-lipid modification S-acylation, the reversible addition of fatty acids to cysteine residues. S-acylation is more commonly referred to as S-palmitoylation or simply palmitoylation, which is the addition of the 16-carbon fatty acid palmitate to proteins. Like phosphorylation, palmitoylation is highly dynamic and tightly regulated by writers (i.e., palmitoyl acyltransferases) and erasers (i.e., depalmitoylating enzymes). The hydrophobic fatty acid anchors proteins to membranes; thus, the reversibility allows proteins to be re-directed to and from membranes based on local signaling factors. This is particularly important in the nervous system, where axons (output projections) can be meters long. Any disturbance in protein trafficking can have dire consequences. Indeed, many proteins involved in neurodegenerative diseases are palmitoylated, and many more have been identified in palmitoyl-proteomic studies. It follows that palmitoyl acyl transferase enzymes have also been implicated in numerous diseases. In addition, palmitoylation can work in concert with cellular mechanisms, like autophagy, to affect cell health and protein modifications, such as acetylation, nitrosylation, and ubiquitination, to affect protein function and turnover. Limited studies have further revealed a sexually dimorphic pattern of protein palmitoylation. Therefore, palmitoylation can have wide-reaching consequences in neurodegenerative diseases.
Collapse
|
41
|
Brown CR, Foster JD. Palmitoylation regulates human serotonin transporter activity, trafficking, and expression and is modulated by escitalopram. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.540092. [PMID: 37214849 PMCID: PMC10197645 DOI: 10.1101/2023.05.09.540092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In the central nervous system, serotonergic signaling modulates sleep, mood, and cognitive control. During neuronal transmission, the synaptic concentration of serotonin is tightly controlled in a spatial and temporal manner by the serotonin transporter (SERT). Dysregulation of serotonergic signaling is implicated in the pathogenesis of major-depressive, obsessive-compulsive, and autism-spectrum disorders, which makes SERT a primary target for prescription therapeutics, most notably selective-serotonin reuptake inhibitors (SSRIs). S-palmitoylation is an increasingly recognized dynamic post-translational modification, regulating protein kinetics, trafficking, and localization patterns upon physiologic/cellular stimuli. In this study, we reveal that human SERTs are a target for palmitoylation, and using the irreversible palmitoyl acyl-transferase inhibitor, 2-bromopalmitate (2BP) we have identified several associated functions. Using a lower dose of 2BP in shorter time frames, inhibition of palmitoylation was associated with reductions in SERT V max , without changes in K m or surface expression. With higher doses of 2BP for longer time intervals, inhibition of palmitoylation was consistent with the loss of cell surface and total SERT protein, suggesting palmitoylation is an important mechanism in regulating SERT trafficking and maintenance of SERT protein through biogenic or anti-degradative processes. Additionally, we have identified that treatment with the SSRI escitalopram decreases SERT palmitoylation analogous to 2BP, reducing SERT surface expression and transport capacity. Ultimately, these results reveal palmitoylation is a major regulatory mechanism for SERT kinetics and trafficking and may be the mechanism responsible for escitalopram-induced internalization and loss of total SERT protein.
Collapse
|
42
|
Koropouli E, Wang Q, Mejías R, Hand R, Wang T, Ginty DD, Kolodkin AL. Palmitoylation regulates neuropilin-2 localization and function in cortical neurons and conveys specificity to semaphorin signaling via palmitoyl acyltransferases. eLife 2023; 12:e83217. [PMID: 37010951 PMCID: PMC10069869 DOI: 10.7554/elife.83217] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 02/22/2023] [Indexed: 04/04/2023] Open
Abstract
Secreted semaphorin 3F (Sema3F) and semaphorin 3A (Sema3A) exhibit remarkably distinct effects on deep layer excitatory cortical pyramidal neurons; Sema3F mediates dendritic spine pruning, whereas Sema3A promotes the elaboration of basal dendrites. Sema3F and Sema3A signal through distinct holoreceptors that include neuropilin-2 (Nrp2)/plexinA3 (PlexA3) and neuropilin-1 (Nrp1)/PlexA4, respectively. We find that Nrp2 and Nrp1 are S-palmitoylated in cortical neurons and that palmitoylation of select Nrp2 cysteines is required for its proper subcellular localization, cell surface clustering, and also for Sema3F/Nrp2-dependent dendritic spine pruning in cortical neurons, both in vitro and in vivo. Moreover, we show that the palmitoyl acyltransferase ZDHHC15 is required for Nrp2 palmitoylation and Sema3F/Nrp2-dependent dendritic spine pruning, but it is dispensable for Nrp1 palmitoylation and Sema3A/Nrp1-dependent basal dendritic elaboration. Therefore, palmitoyl acyltransferase-substrate specificity is essential for establishing compartmentalized neuronal structure and functional responses to extrinsic guidance cues.
Collapse
Affiliation(s)
- Eleftheria Koropouli
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Qiang Wang
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Rebeca Mejías
- Department of Physiology,University of SevilleSevilleSpain
| | - Randal Hand
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Tao Wang
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Alex L Kolodkin
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
43
|
Casellas-Vidal D, Mademont-Soler I, Sánchez J, Plaja A, Castells N, Camós M, Nieto-Moragas J, Del Mar García M, Rodriguez-Solera C, Rivera H, Brunet J, Álvarez S, Perapoch J, Queralt X, Obón M. ZDHHC15 as a candidate gene for autism spectrum disorder. Am J Med Genet A 2023; 191:941-947. [PMID: 36565021 DOI: 10.1002/ajmg.a.63099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
The phenotypic repercussion of ZDHHC15 haploinsufficiency is not well-known. This gene was initially suggested as a candidate for X-linked mental retardation, but such an association was later questioned. We studied a multiplex family with three members with autism spectrum disorder (ASD) by array CGH, karyotype, exome sequencing and X-chromosome inactivation patterns. Medical history interviews, cognitive and physical examinations, and sensory profiling were also assessed. The three family members with ASD (with normal cognitive abilities and an abnormal sensory profile) were the only carriers of a 1.7 Mb deletion in the long arm of chromosome X, involving: ZDHHC15, MAGEE2, PBDC1, MAGEE1, MIR384 and MIR325. The normal chromosome X was preferentially inactivated in female carriers, and the whole exome sequencing of an affected family member did not reveal any additional genetic variant that could explain the phenotype. Thus, in the present family, ASD segregates with a deletion on chromosome X that includes ZDHHC15. Considering our results together with gene data (regarding function, expression, conservation and animal/cellular models), ZDHHC15 is a candidate gene for ASD. Emerging evidence also suggests that this gene could be associated with other neurodevelopmental disorders, with incomplete penetrance and variable expressivity.
Collapse
Affiliation(s)
| | - Irene Mademont-Soler
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| | - Joana Sánchez
- Centre de Salut Mental Infantil i Juvenil, Institut d'Assistència Sanitària, Girona, Spain
| | - Alberto Plaja
- Unitat d'Arrays, Departament de Genètica, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Neus Castells
- Unitat d'Arrays, Departament de Genètica, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Maria Camós
- Servei de Pediatria, Hospital Universitari Doctor Josep Trueta, Girona, Spain
| | - Javier Nieto-Moragas
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| | | | | | - Helena Rivera
- Centre de Salut Mental Infantil i Juvenil, Institut d'Assistència Sanitària, Girona, Spain
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology, Hospital Universitari Doctor Josep Trueta, IDIBGI, Girona, Spain
| | - Sara Álvarez
- Servicio de Diagnóstico Genético, NIMGenetics, Madrid, Spain
| | - Josep Perapoch
- Servei de Pediatria, Hospital Universitari Doctor Josep Trueta, Girona, Spain
| | - Xavier Queralt
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| | - María Obón
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial Girona, Institut Català de la Salut, Girona, Spain
| |
Collapse
|
44
|
Gupta JP, Jenkins PM. Ankyrin-B is lipid-modified by S-palmitoylation to promote dendritic membrane scaffolding of voltage-gated sodium channel Na V1.2 in neurons. Front Physiol 2023; 14:959660. [PMID: 37064897 PMCID: PMC10098127 DOI: 10.3389/fphys.2023.959660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 03/21/2023] [Indexed: 04/01/2023] Open
Abstract
Neuronal ankyrin-B is an intracellular scaffolding protein that plays multiple roles in the axon. By contrast, relatively little is known about the function of ankyrin-B in dendrites, where ankyrin-B is also localized in mature neurons. Recently, we showed that ankyrin-B acts as a scaffold for the voltage-gated sodium channel, NaV1.2, in dendrites of neocortical pyramidal neurons. How ankyrin-B is itself targeted to the dendritic membrane is not well understood. Here, we report that ankyrin-B is lipid-modified by S-palmitoylation to promote dendritic localization of NaV1.2. We identify the palmitoyl acyl transferase zDHHC17 as a key mediator of ankyrin-B palmitoylation in heterologous cells and in neurons. Additionally, we find that zDHHC17 regulates ankyrin-B protein levels independently of its S-acylation function through a conserved binding mechanism between the ANK repeat domain of zDHHC17 and the zDHHC ankyrin-repeat binding motif of ankyrin-B. We subsequently identify five cysteines in the N-terminal ankyrin repeat domain of ankyrin-B that are necessary for ankyrin-B palmitoylation. Mutation of these five cysteines to alanines not only abolishes ankyrin-B palmitoylation, but also prevents ankyrin-B from scaffolding NaV1.2 at dendritic membranes of neurons due to ankyrin-B's inability to localize properly at dendrites. Thus, we show palmitoylation is critical for localization and function of ankyrin-B at dendrites. Strikingly, loss of ankyrin-B palmitoylation does not affect ankyrin-B-mediated axonal cargo transport of synaptic vesicle synaptotagmin-1 in neurons. This is the first demonstration of S-palmitoylation of ankyrin-B as an underlying mechanism required for ankyrin-B localization and function in scaffolding NaV1.2 at dendrites.
Collapse
Affiliation(s)
- Julie P. Gupta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Paul M. Jenkins
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
45
|
Inhibition of hippocampal palmitoyl acyltransferase activity impairs spatial learning and memory consolidation. Neurobiol Learn Mem 2023; 200:107733. [PMID: 36804592 DOI: 10.1016/j.nlm.2023.107733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/12/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Protein palmitoylation regulates trafficking, mobilization, localization, interaction, and distribution of proteins through the palmitoyl acyltransferases (PATs) enzymes. Protein palmitoylation controls rapid and dynamic changes of the synaptic architecture that modifies the efficiency and strength of synaptic connections, a fundamental mechanism to generate stable and long-lasting memory traces. Although protein palmitoylation in functional synaptic plasticity has been widely described, its role in learning and memory processes is poorly understood. In this work, we found that PATs inhibition into the hippocampus before and after the training of Morris water maze (MWM) and object location memory (OLM) impaired spatial learning. However, we demonstrated that PATs inhibition during the retrieval does not affect the expression of spatial memory in both MWM and OLM. Accordingly, long-term potentiation induction is impaired by inhibiting PATs into the hippocampus before high-frequency electrical stimulation but not after. These findings suggest that PATs activity is necessary to modify neural plasticity, a mechanism required for memory acquisition and consolidation. Like phosphorylation, active palmitoylation is required to regulate the function of already existing proteins that change synaptic strength in the hippocampus to acquire and later consolidate spatial memories.
Collapse
|
46
|
Niki Y, Adachi N, Fukata M, Fukata Y, Oku S, Makino-Okamura C, Takeuchi S, Wakamatsu K, Ito S, Declercq L, Yarosh DB, Mammone T, Nishigori C, Saito N, Ueyama T. S-Palmitoylation of Tyrosinase at Cysteine 500 Regulates Melanogenesis. J Invest Dermatol 2023; 143:317-327.e6. [PMID: 36063887 DOI: 10.1016/j.jid.2022.08.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 01/25/2023]
Abstract
Palmitoylation is a lipid modification involving the attachment of palmitic acid to a cysteine residue, thereby affecting protein function. We investigated the effect of palmitoylation of tyrosinase, the rate-limiting enzyme in melanin synthesis, using a human three-dimensional skin model system and melanocyte culture. The palmitoylation inhibitor, 2-bromopalmitate, increased melanin content and tyrosinase protein levels in melanogenic cells by suppressing tyrosinase degradation. The palmitoylation site was Cysteine500 in the C-terminal cytoplasmic tail of tyrosinase. The nonpalmitoylatable mutant, tyrosinase (C500A), was slowly degraded and less ubiquitinated than wild-type tyrosinase. Screening for the Asp-His-His-Cys (DHHC) family of proteins for tyrosinase palmitoylation suggested that DHHC2, 3, 7, and 15 are involved in tyrosinase palmitoylation. Knockdown of DHHC2, 3, or 15 increased tyrosinase protein levels and melanin content. Determination of their subcellular localization in primary melanocytes revealed that DHHC2, 3, and 15 were localized in the endoplasmic reticulum, Golgi apparatus, and/or melanosomes, whereas only DHHC2 was localized in the melanosomes. Immunoprecipitation showed that DHHC2 and DHHC3 predominantly bind to mature and immature tyrosinase, respectively. Taken together, tyrosinase palmitoylation at Cysteine500 by DHHC2, 3, and/or 15, especially DHHC2 in trans-Golgi apparatus and melanosomes and DHHC3 in the endoplasmic reticulum and cis-Golgi apparatus, regulate melanogenesis by modulating tyrosinase protein levels.
Collapse
Affiliation(s)
- Yoko Niki
- Kobe Skin Research Department, Biosignal Research Center, Kobe University, Kobe, Japan; School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Japan
| | - Naoko Adachi
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Shinichiro Oku
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Chieko Makino-Okamura
- Kobe Skin Research Department, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Seiji Takeuchi
- Kobe Skin Research Department, Biosignal Research Center, Kobe University, Kobe, Japan; Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, Kobe, Japan
| | | | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Aichi, Japan
| | - Lieve Declercq
- Research & Development, Estee Lauder Companies, Melville, New York, USA
| | - Daniel B Yarosh
- Research & Development, Estee Lauder Companies, Melville, New York, USA
| | - Tomas Mammone
- Research & Development, Estee Lauder Companies, Melville, New York, USA
| | - Chikako Nishigori
- Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Naoaki Saito
- Kobe Skin Research Department, Biosignal Research Center, Kobe University, Kobe, Japan; Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan.
| |
Collapse
|
47
|
Buszka A, Pytyś A, Colvin D, Włodarczyk J, Wójtowicz T. S-Palmitoylation of Synaptic Proteins in Neuronal Plasticity in Normal and Pathological Brains. Cells 2023; 12:cells12030387. [PMID: 36766729 PMCID: PMC9913408 DOI: 10.3390/cells12030387] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/08/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Protein lipidation is a common post-translational modification of proteins that plays an important role in human physiology and pathology. One form of protein lipidation, S-palmitoylation, involves the addition of a 16-carbon fatty acid (palmitate) onto proteins. This reversible modification may affect the regulation of protein trafficking and stability in membranes. From multiple recent experimental studies, a picture emerges whereby protein S-palmitoylation is a ubiquitous yet discrete molecular switch enabling the expansion of protein functions and subcellular localization in minutes to hours. Neural tissue is particularly rich in proteins that are regulated by S-palmitoylation. A surge of novel methods of detection of protein lipidation at high resolution allowed us to get better insights into the roles of protein palmitoylation in brain physiology and pathophysiology. In this review, we specifically discuss experimental work devoted to understanding the impact of protein palmitoylation on functional changes in the excitatory and inhibitory synapses associated with neuronal activity and neuronal plasticity. The accumulated evidence also implies a crucial role of S-palmitoylation in learning and memory, and brain disorders associated with impaired cognitive functions.
Collapse
|
48
|
Komaniecki G, Camarena MDC, Gelsleichter E, Mendoza R, Subler M, Windle JJ, Dozmorov MG, Lai Z, Sarkar D, Lin H. Astrocyte Elevated Gene-1 Cys75 S-Palmitoylation by ZDHHC6 Regulates Its Biological Activity. Biochemistry 2023; 62:543-553. [PMID: 36548985 PMCID: PMC9850907 DOI: 10.1021/acs.biochem.2c00583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Nonalcoholic fatty liver disease is a major risk factor for hepatocellular carcinoma (HCC). Astrocyte elevated gene-1/Metadherin (AEG-1/MTDH) augments lipid accumulation (steatosis), inflammation, and tumorigenesis, thereby promoting the whole spectrum of this disease process. Targeting AEG-1 is a potential interventional strategy for nonalcoholic steatohepatitis (NASH) and HCC. Thus, proper understanding of the regulation of this molecule is essential. We found that AEG-1 is palmitoylated at residue cysteine 75 (Cys75). Mutation of Cys75 to serine (Ser) completely abolished AEG-1 palmitoylation. We identified ZDHHC6 as a palmitoyltransferase catalyzing the process in HEK293T cells. To obtain insight into how palmitoylation regulates AEG-1 function, we generated knock-in mice by CRISPR/Cas9 in which Cys75 of AEG-1 was mutated to Ser (AEG-1-C75S). No developmental or anatomical abnormality was observed between AEG-1-wild type (AEG-1-WT) and AEG-1-C75S littermates. However, global gene expression analysis by RNA-sequencing unraveled that signaling pathways and upstream regulators, which contribute to cell proliferation, motility, inflammation, angiogenesis, and lipid accumulation, were activated in AEG-1-C75S hepatocytes compared to AEG-1-WT. These findings suggest that AEG-1-C75S functions as dominant positive and that palmitoylation restricts oncogenic and NASH-promoting functions of AEG-1. We thus identify a previously unknown regulatory mechanism of AEG-1, which might help design new therapeutic strategies for NASH and HCC.
Collapse
Affiliation(s)
- Garrison Komaniecki
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
- C.
Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Maria Del Carmen Camarena
- C.
Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Eric Gelsleichter
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Rachel Mendoza
- Department
of Human and Molecular Genetics, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Mark Subler
- Department
of Human and Molecular Genetics, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Jolene J. Windle
- Department
of Human and Molecular Genetics, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
- Massey
Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- VCU
Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Mikhail G. Dozmorov
- Department
of Biostatistics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
- Department
of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Zhao Lai
- Greehy
Children’s Cancer Research Institute, University of Texas Health
Science Center San Antonio, San Antonio, Texas 78229, United States
| | - Devanand Sarkar
- Department
of Human and Molecular Genetics, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
- Massey
Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- VCU
Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Hening Lin
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
- Howard
Hughes Medical Institute, Department of Chemistry and Chemical Biology,
Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
49
|
Butler L, Locatelli C, Allagioti D, Lousa I, Lemonidis K, Tomkinson NCO, Salaun C, Chamberlain LH. S-acylation of Sprouty and SPRED proteins by the S-acyltransferase zDHHC17 involves a novel mode of enzyme-substrate interaction. J Biol Chem 2023; 299:102754. [PMID: 36442513 PMCID: PMC9800311 DOI: 10.1016/j.jbc.2022.102754] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
S-acylation is an essential post-translational modification, which is mediated by a family of 23 zDHHC enzymes in humans. Several thousand proteins are modified by S-acylation; however, we lack a detailed understanding of how enzyme-substrate recognition and specificity is achieved. Previous work showed that the ankyrin repeat domain of zDHHC17 (ANK17) recognizes a short linear motif, known as the zDHHC ANK binding motif (zDABM) in substrate protein SNAP25, as a mechanism of substrate recruitment prior to S-acylation. Here, we investigated the S-acylation of the Sprouty and SPRED family of proteins by zDHHC17. Interestingly, although Sprouty-2 (Spry2) contains a zDABM that interacts with ANK17, this mode of binding is dispensable for S-acylation, and indeed removal of the zDABM does not completely ablate binding to zDHHC17. Furthermore, the related SPRED3 protein interacts with and is efficiently S-acylated by zDHHC17, despite lacking a zDABM. We undertook mutational analysis of SPRED3 to better understand the basis of its zDABM-independent interaction with zDHHC17. This analysis found that the cysteine-rich SPR domain of SPRED3, which is the defining feature of all Sprouty and SPRED proteins, interacts with zDHHC17. Surprisingly, the interaction with SPRED3 was independent of ANK17. Our mutational analysis of Spry2 was consistent with the SPR domain of this protein containing a zDHHC17-binding site, and Spry2 also showed detectable binding to a zDHHC17 mutant lacking the ANK domain. Thus, zDHHC17 can recognize its substrates through zDABM-dependent and/or zDABM-independent mechanisms, and some substrates display more than one mode of binding to this enzyme.
Collapse
Affiliation(s)
- Liam Butler
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Carolina Locatelli
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Despoina Allagioti
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Irina Lousa
- UCIBIO, REQUIMTE, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Kimon Lemonidis
- School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicholas C O Tomkinson
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Christine Salaun
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Luke H Chamberlain
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom.
| |
Collapse
|
50
|
Pei X, Li KY, Shen Y, Li JT, Lei MZ, Fang CY, Lu HJ, Yang HJ, Wen W, Yin M, Qu J, Lei QY. Palmitoylation of MDH2 by ZDHHC18 activates mitochondrial respiration and accelerates ovarian cancer growth. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2017-2030. [PMID: 35366151 DOI: 10.1007/s11427-021-2048-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022]
Abstract
Epithelial ovarian cancer (EOC) exhibits strong dependency on the tricarboxylic acid (TCA) cycle and oxidative phosphorylation to fuel anabolic process. Here, we show that malate dehydrogenase 2 (MDH2), a key enzyme of the TCA cycle, is palmitoylated at cysteine 138 (C138) residue, resulting in increased activity of MDH2. We next identify that ZDHHC18 acts as a palmitoyltransferase of MDH2. Glutamine deprivation enhances MDH2 palmitoylation by increasing the binding between ZDHHC18 and MDH2. MDH2 silencing represses mitochondrial respiration as well as ovarian cancer cell proliferation both in vitro and in vivo. Intriguingly, re-expression of wild-type MDH2, but not its palmitoylation-deficient C138S mutant, sustains mitochondrial respiration and restores the growth as well as clonogenic capability of ovarian cancer cells. Notably, MDH2 palmitoylation level is elevated in clinical cancer samples from patients with high-grade serous ovarian cancer. These observations suggest that MDH2 palmitoylation catalyzed by ZDHHC18 sustains mitochondrial respiration and promotes the malignancy of ovarian cancer, yielding possibilities of targeting ZDHHC18-mediated MDH2 palmitoylation in the treatment of EOC.
Collapse
Affiliation(s)
- Xuan Pei
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Kai-Yue Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuan Shen
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jin-Tao Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ming-Zhu Lei
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cai-Yun Fang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hao-Jie Lu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hui-Juan Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Wenyu Wen
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Miao Yin
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jia Qu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Qun-Ying Lei
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.
| |
Collapse
|