1
|
Cao H, Gao H, Li Y, Li L, Liu S, Jin T, Wang Y, Gong Y, Yuan S, Dong W. Zinc finger DHHC-type palmitoyltransferase 13-mediated S-palmitoylation of GNA13 from Sertoli cell-derived extracellular vesicles inhibits autophagy in spermatogonial stem cells. Cell Commun Signal 2025; 23:178. [PMID: 40205436 PMCID: PMC11983822 DOI: 10.1186/s12964-025-02177-0] [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: 01/15/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Extracellular vesicles (EVs) originating from testicular somatic cells act as pivotal intermediaries in cell signaling crosstalk between spermatogenic cells and the testicular microenvironment. The intricate balance between palmitoylation and depalmitoylation governs the positioning of protein cargos on the membrane, thereby influencing cellular activities by concentrating these proteins in EVs for delivery to recipient cells. Here, we reveal that GNA13 undergoes specific S-palmitoylation at Cys14 and Cys18 residues in Sertoli cells (SCs), a modification essential for its localization to the plasma membrane. We identify DHHC13, a member of the zinc finger DHHC-type palmitoyltransferase family that catalyzes protein S-palmitoylation, as the enzyme responsible for this critical post-translational modification. Additionally, GNA13 palmitoylation is indispensable for its selective enrichment in EVs emanating from SCs. Intriguingly, we discovered the presence of palmitoylated GNA13 in SC-derived EVs significantly downregulates autophagy levels in spermatogonial stem cells (SSCs), and the inhibition of GNA13 palmitoylation attenuates its interaction with ARHGEF12 which leads to diminished RhoA activity and consequent elevation of autophagy in SSCs. Our results illuminate the crucial role of DHHC13-mediated GNA13 S-palmitoylation in modulating autophagy levels in SSCs through SCs-derived EVs, suggesting that PM-GNA13-EV may serve as a potential candidate for further exploration in addressing fertility-related challenges during spermatogenesis.
Collapse
Affiliation(s)
- Heran Cao
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Huihui Gao
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Li
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Tianqi Jin
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Yang Wang
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Ye Gong
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
2
|
Holme S, Sapia J, Davey M, Vanni S, Conibear E. An S-acylated N-terminus and a conserved loop regulate the activity of the ABHD17 deacylase. J Cell Biol 2025; 224:e202405042. [PMID: 39951021 PMCID: PMC11827582 DOI: 10.1083/jcb.202405042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/07/2024] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
The dynamic addition and removal of long-chain fatty acids modulate protein function and localization. The α/β hydrolase domain-containing (ABHD) 17 enzymes remove acyl chains from membrane-localized proteins such as the oncoprotein NRas, but how the ABHD17 proteins are regulated is unknown. Here, we used cell-based studies and molecular dynamics simulations to show that ABHD17 activity is controlled by two mobile elements-an S-acylated N-terminal helix and a loop-that flank the putative substrate-binding pocket. Multiple S-acylation events anchor the N-terminal helix in the membrane, enabling hydrophobic residues in the loop to engage with the bilayer. This stabilizes the conformation of both helix and loop, alters the conformation of the binding pocket, and optimally positions the enzyme for substrate engagement. S-acylation may be a general feature of acyl-protein thioesterases. By providing a mechanistic understanding of how the lipid modification of a lipid-removing enzyme promotes its enzymatic activity, this work contributes to our understanding of cellular S-acylation cycles.
Collapse
Affiliation(s)
- Sydney Holme
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer Sapia
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Michael Davey
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Swiss National Center for Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Elizabeth Conibear
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Zou G, Tang Y, Yang J, Fu S, Li Y, Ren X, Zhou N, Zhao W, Gao J, Ruan Z, Jiang Z. Signal-induced NLRP3 phase separation initiates inflammasome activation. Cell Res 2025:10.1038/s41422-025-01096-6. [PMID: 40164768 DOI: 10.1038/s41422-025-01096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
NLRP3 inflammasome is activated by diverse stimuli including infections, intracellular and environmental irritants. How NLRP3 senses these unrelated stimuli and what activates NLRP3 remain unknown. Here we report that signal-dependent NLRP3 phase separation initiated its activation, in which the palmitoyltransferase ZDHHC7-mediated tonic NLRP3 palmitoylation and an IDR region in the FISNA domain of NLRP3 play important roles. Moreover, three conserved hydrophobic residues in the IDR critically mediate multivalent weak interactions. NLRP3-activating stimuli including K+ efflux and NLRP3-interacting molecules imiquimod, palmitate, and cardiolipin all cause NLRP3 conformational change and induce its phase separation and activation in cells and/or in vitro. Surprisingly, amphiphilic molecules like di-alcohols used to inhibit biomolecular phase separation and chemotherapeutic drugs doxorubicin and paclitaxel activate NLRP3 independently of ZDHHC7 by directly inducing NLRP3 phase separation. Mechanistically, amphiphilic molecules decrease the solubility of both palmitoylated and non-palmitoylated NLRP3 to directly induce its phase separation and activation while NLRP3 palmitoylation reduces its solubility to some extent without activation. Therefore, ZDHHC7-mediated NLRP3 palmitoylation in resting cells licenses its activation by lowering the threshold for NLRP3 phase separation in response to any of the diverse stimuli whereas NLRP3 solubility-reducing molecules like di-alcohols and chemotherapeutic drugs activate NLRP3 directly. The signal-induced NLRP3 phase separation likely provides the simplest and most direct mechanistic basis for NLRP3 activation.
Collapse
Affiliation(s)
- Gonglu Zou
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yuluan Tang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jie Yang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Shuo Fu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yuheng Li
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xuanyao Ren
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Nanhai Zhou
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Wenlong Zhao
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Juyi Gao
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Ziran Ruan
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
4
|
Li Y, Liu Y, Wang C. Quantitative profiling of PTM stoichiometry by DNA mass tags. Bioorg Med Chem 2025; 118:118050. [PMID: 39724823 DOI: 10.1016/j.bmc.2024.118050] [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: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Protein post-translational modification (PTM) serves as an important mechanism for regulating protein function. Accurate assay of PTM stoichiometry, or PTM occupancy, which refers to the proportion of proteins that contain specific modifications, is important for understanding the function of PTMs. We previously developed a novel chemoproteomic strategy "STO-MS" to quantify the PTM stoichiometry in complex biological samples, which employs a resolvable polymer mass tag to differentiate modified proteins and utilizes liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) techniques to measure PTM stoichiometry. However, the resolution of STO-MS is constrained by the relatively low molecular weight of the mass tag, and the incorporation of isotopic labels not only complicates the sample preparation but also restricts the measurement throughput. To address these challenges, we herein developed "STO-MS+", an enhanced workflow, that incorporates an optimized DNA mass tag and employs a label-free quantitative data analysis approach. We applied STO-MS+ to measure stoichiometry of three distinct PTMs, including endogenous carbonylation induced by arachidonic acid (AA), itaconation, and endogenous O-GlcNAcylation. Our work marks a notable improvement in chemoproteomic methodologies for quantifying post-translational modifications and provides a powerful analytical tool for PTM research.
Collapse
Affiliation(s)
- Yuanpei Li
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuan Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Chu Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| |
Collapse
|
5
|
Wang J, Shen D, Jiang J, Hu L, Fang K, Xie C, Shen N, Zhou Y, Wang Y, Du S, Meng S. Dietary Palmitic Acid Drives a Palmitoyltransferase ZDHHC15-YAP Feedback Loop Promoting Tumor Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409883. [PMID: 39686664 PMCID: PMC11809420 DOI: 10.1002/advs.202409883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Indexed: 12/18/2024]
Abstract
Elevated uptake of saturated fatty acid palmitic acid (PA) is associated with tumor metastasis; however, the precise mechanisms remain partially understood, hindering the development of therapy for PA-driven tumor metastasis. The Hippo-Yes-associated protein (Hippo/YAP) pathway is implicated in cancer progression. Here it is shown that a high-palm oil diet potentiates tumor metastasis in murine xenografts in part through YAP. It is found that the palmitoyltransferase ZDHHC15 is a YAP-regulated gene that forms a feedback loop with YAP. Notably, PA drives the ZDHHC15-YAP feedback loop, thus enforces YAP signaling, and hence promotes tumor metastasis in murine xenografts. In addition, it is shown that ZDHHC15 associates with Kidney and brain protein (KIBRA, also known as WW- and C2 domain-containing protein 1, WWC1), an upstream component of Hippo signaling, and mediates its palmitoylation. KIBRA palmitoylation leads to its degradation and regulates its subcellular localization and activity toward the Hippo/YAP pathway. Moreover, PA enhances KIBRA palmitoylation and degradation. It is further shown that combinatorial targeting of YAP and fatty acid synthesis exhibits augmented effects against metastasis formation in mice fed with a Palm diet. Collectively, these findings uncover a ZDHHC15-YAP feedback loop as a previously unrecognized mechanism underlying PA-promoted tumor metastasis and support targeting YAP and fatty acid synthesis as potential therapeutic targets in PA-driven tumor metastasis.
Collapse
Affiliation(s)
- Jianxin Wang
- Institute of Cancer Stem CellDalian Medical University Cancer CenterDalian116044China
| | - Dachuan Shen
- Department of OncologyAffiliated Zhongshan Hospital of Dalian UniversityDalian116001China
| | - Jian Jiang
- Central Hospital of Dalian University of TechnologyDepartment of Spine SurgeryDalian116033China
| | - Lulu Hu
- Department of Laboratory MedicineQingdao Central HospitalUniversity of Health and Rehabilitation Sciences NO.369Dengyun Road, Qingdao National High‐tech Industrial Development ZoneQingdaoChina
| | - Kun Fang
- Central LaboratoryCancer Hospital of China Medical UniversityCancer Hospital of Dalian University of TechnologyLiaoning Cancer Hospital & InstituteShenyang110042China
| | - Chunrui Xie
- Institute of Cancer Stem CellDalian Medical University Cancer CenterDalian116044China
| | - Ning Shen
- Institute of Cancer Stem CellDalian Medical University Cancer CenterDalian116044China
| | - Yuzhao Zhou
- Institute of Cancer Stem CellDalian Medical University Cancer CenterDalian116044China
| | - Yifei Wang
- Department of Obstetrics and GynecologyAffiliated Zhongshan Hospital of Dalian UniversityDalian116001China
| | - Sha Du
- Institute of Cancer Stem CellDalian Medical University Cancer CenterDalian116044China
| | - Songshu Meng
- Institute of Cancer Stem CellDalian Medical University Cancer CenterDalian116044China
| |
Collapse
|
6
|
Shi X, Shen L, Chen S, Liu M, Wang J, Wen X, Liu W, Mao L, Ding Y, Yu L, Xu J. Swine RNF5 positively regulates the antiviral activity of IFITM1 by mediating the degradation of ABHD16A. J Virol 2025; 99:e0127724. [PMID: 39601593 PMCID: PMC11784460 DOI: 10.1128/jvi.01277-24] [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: 07/22/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Interferon-inducible transmembrane (IFITM) proteins are broad-spectrum antiviral factors that confer cellular resistance to virus invasion. α/β-Hydrolase domain-containing 16A (ABHD16A) has recently been identified as a novel depalmitoylase that can inhibit the antiviral activity of IFITM proteins by catalyzing the depalmitoyl reaction; this pattern may be crucial for the host to avoid damage caused by excessive immune response. However, it remains largely elusive about how host cells regulate the activity of ABHD16A. In the present study, we performed the AlphaFold2-based protein-protein interaction prediction and identified swine E3 ubiquitin ligase ring finger protein 5 (sRNF5) as a sABHD16A-interacting protein and negatively regulated the stability of sABHD16A. Using immunofluorescence and co-immunoprecipitation techniques, we uncovered that sRNF5 targeted sABHD16A for ubiquitination and degradation via the proteasomal pathway at residues K3 and K452. Furthermore, sABHD16A catalyzed the depalmitoylation of sIFITM1, which obstructed the antiviral function of sIFITM1, while sRNF5 caused ubiquitination of sABHD16A, which attenuated the depalmitoylation effect on sIFITM1, and consequently restored the antiviral activity of sIFITM1. Collectively, our findings demonstrate for the first time that sRNF5 positively regulates the antiviral function of sIFITM1 by mediating the degradation of sABHD16A, which expands the biological functions of RNF5 and ABHD16A in immune regulation. Moreover, our work highlights the well-designed interplay between RNF5, ABHD16A, and IFITM, which balances antiviral immune responses to avoid the disorders induced by excessive immune response. IMPORTANCE Interferon and interferon-stimulated genes play significant and protective roles in the host's defense against viral infection. IFITM family proteins, which can be strongly induced by interferon, have been identified as the first line of defense to prevent invasion of various viruses. Further analysis reveals the antiviral activity of IFITMs depends on palmitoylation/depalmitoylation. Recently, we reported that ABHD16A, as the first depalmitoylase of IFITMs, negatively regulated the antiviral activity of IFITMs. However, these raise crucial questions: how ABHD16A is regulated and remained in a balanced manner? Here, we show that swine RNF5 attenuates the negative regulation of sIFITM1 against virus invasion by modifying sABHD16A through ubiquitination and guiding sABHD16A for degradation. Thus, sRNF5-sABHD16A interplay plays an indispensable role in regulating immune response and avoiding the disorders induced by elevated interferon levels. Overall, our findings extend the upstream subtle regulatory molecular mechanism of IFITMs and provide potential targets for viral disease therapy.
Collapse
Affiliation(s)
- Xuemeng Shi
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Lingyi Shen
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Shuaiwu Chen
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Mingyang Liu
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Jingyi Wang
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Xin Wen
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Wei Liu
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Lin Mao
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yunyun Ding
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Li Yu
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| | - Jun Xu
- College of Life Science, Zhengdong New District Longzi Lake Campus, Henan Agricultural University, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Bradburn DA, Reis JC, Qayyum S, Viennet T, Arthanari H, Cyert MS. A novel motif in calcimembrin/C16orf74 dictates multimeric dephosphorylation by calcineurin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.12.593783. [PMID: 38798520 PMCID: PMC11118366 DOI: 10.1101/2024.05.12.593783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Calcineurin, the Ca 2+ /calmodulin-activated protein phosphatase, recognizes substrates and regulators via short linear motifs, PxIxIT and LxVP, which dock to distinct sites on calcineurin and determine calcineurin distribution and catalysis, respectively. Calcimembrin/C16orf74 (CLMB), an intrinsically disordered microprotein whose expression correlates with poor cancer outcomes, targets calcineurin to membranes where it may promote oncogenesis by shaping calcineurin signaling. We show that CLMB associates with membranes via lipidation, i.e. N-myristoylation and reversible S-acylation. Furthermore, CLMB contains an unusual composite 'LxVPxIxIT' motif, that binds the PxIxIT-docking site on calcineurin with extraordinarily high affinity when phosphorylated, 33 LxVPxIxITxx(p)T 44 . Calcineurin dephosphorylates CLMB to decrease this affinity, but Thr44 is protected from dephosphorylation when PxIxIT-bound. We propose that CLMB is dephosphorylated in multimeric complexes, where one PxIxIT-bound CLMB recruits calcineurin to membranes, allowing a second CLMB to engage via its LxVP motif to be dephosphorylated. In vivo and in vitro data, including nuclear magnetic resonance (NMR) analyses of CLMB-calcineurin complexes, supports this model. Thus, the CLMB composite motif imposes unique properties to calcineurin signaling at membranes including sensitivity to CLMB:calcineurin ratios, CLMB phosphorylation and dynamic S-acylation.
Collapse
|
8
|
Gu S, Nie X, George A, Tyler K, Xing Y, Qin L, Qi B. Bioinformatics and Expression Profiling of the DHHC-CRD S-Acyltransferases Reveal Their Roles in Growth and Stress Response in Woodland Strawberry ( Fragaria vesca). PLANTS (BASEL, SWITZERLAND) 2025; 14:127. [PMID: 39795387 PMCID: PMC11722789 DOI: 10.3390/plants14010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025]
Abstract
Protein S-acyl transferases (PATs) are a family of enzymes that catalyze protein S-acylation, a post-translational lipid modification involved in protein membrane targeting, trafficking, stability, and protein-protein interaction. S-acylation plays important roles in plant growth, development, and stress responses. Here, we report the genome-wide analysis of the PAT family genes in the woodland strawberry (Fragaria vesca), a model plant for studying the economically important Rosaceae family. In total, 21 'Asp-His-His-Cys' Cys Rich Domain (DHHC-CRD)-containing sequences were identified, named here as FvPAT1-21. Expression profiling by reverse transcription quantitative PCR (RT-qPCR) showed that all the 21 FvPATs were expressed ubiquitously in seedlings and different tissues from adult plants, with notably high levels present in vegetative tissues and young fruits. Treating seedlings with hormones indole-3-acetic acid (IAA), abscisic acid (ABA), and salicylic acid (SA) rapidly increased the transcription of most FvPATs. A complementation assay in yeast PAT mutant akr1 and auto-S-acylation assay of one FvPAT (FvPAT19) confirmed its enzyme activity where the Cys in the DHHC motif was required. An AlphaFold prediction of the DHHC and the mutated DHHC155S of FvPAT19 provided further proof of the importance of C155 in fatty acid binding. Together, our data clearly demonstrated that S-acylation catalyzed by FvPATs plays important roles in growth, development, and stress signaling in strawberries. These preliminary results could contribute to further research to understand S-acylation in strawberries and plants in general.
Collapse
Affiliation(s)
- Si Gu
- School of Pharmacy and BioMolecular Sciences, Liverpool John Moores University, Byram Street, Liverpool L3 3AF, UK; (S.G.); (A.G.); (K.T.)
| | - Xinghua Nie
- College of Plant Science and Technology, Beijing University of Agriculture, Beijing 102208, China; (X.N.); (Y.X.); (L.Q.)
| | - Amal George
- School of Pharmacy and BioMolecular Sciences, Liverpool John Moores University, Byram Street, Liverpool L3 3AF, UK; (S.G.); (A.G.); (K.T.)
| | - Kyle Tyler
- School of Pharmacy and BioMolecular Sciences, Liverpool John Moores University, Byram Street, Liverpool L3 3AF, UK; (S.G.); (A.G.); (K.T.)
| | - Yu Xing
- College of Plant Science and Technology, Beijing University of Agriculture, Beijing 102208, China; (X.N.); (Y.X.); (L.Q.)
| | - Ling Qin
- College of Plant Science and Technology, Beijing University of Agriculture, Beijing 102208, China; (X.N.); (Y.X.); (L.Q.)
| | - Baoxiu Qi
- School of Pharmacy and BioMolecular Sciences, Liverpool John Moores University, Byram Street, Liverpool L3 3AF, UK; (S.G.); (A.G.); (K.T.)
| |
Collapse
|
9
|
Chaturvedi S, Sonawane A. Recapitulating the potential contribution of protein S-palmitoylation in cancer. Cancer Metastasis Rev 2024; 44:20. [PMID: 39725785 DOI: 10.1007/s10555-024-10217-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024]
Abstract
Protein S-palmitoylation is a reversible form of protein lipidation in which the formation of a thioester bond occurs between a cysteine (Cys) residue of a protein and a 16-carbon fatty acid chain. This modification is catalyzed by a family of palmitoyl acyl transferases, the DHHC enzymes, so called because of their Asp-His-His-Cys (DHHC) catalytic motif. Deregulation of DHHC enzymes has been linked to various diseases, including cancer and infections. Cancer, a major cause of global mortality, is characterized by features like uncontrolled cell growth, resistance to cell death, angiogenesis, invasion, and metastasis. Several of these processes are controlled by DHHC-mediated S-palmitoylation of oncogenes or tumor suppressors, including growth factor receptors (e.g., EGFR), kinases (e.g., AKT), and transcription factors (e.g., β-catenin). Dynamic regulation of S-palmitoylation is also governed by protein depalmitoylases. These enzymes balance the cycling of palmitoylation and regulate cellular signaling, cell growth, and its organization. Given the significance of S-palmitoylation in cancer, the DHHCs and protein depalmitoylases are promising targets for cancer therapy. Here we summarize the catalytic mechanisms of DHHC enzymes and depalmitoylases, their role in cancer progression and prevention, as well as the crosstalk of palmitoylation with other post-translational modifications. Additionally, we discuss the methods to detect S-palmitoylation, the limitations of available DHHC-targeting inhibitors, and ongoing research efforts to address these obstacles.
Collapse
Affiliation(s)
- Suchi Chaturvedi
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, 453552, Simrol, Madhya Pradesh, India
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, 453552, Simrol, Madhya Pradesh, India.
| |
Collapse
|
10
|
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
|
11
|
Ocasio CA, Baggelaar MP, Sipthorp J, Losada de la Lastra A, Tavares M, Volarić J, Soudy C, Storck EM, Houghton JW, Palma-Duran SA, MacRae JI, Tomić G, Carr L, Downward J, Eggert US, Tate EW. A palmitoyl transferase chemical-genetic system to map ZDHHC-specific S-acylation. Nat Biotechnol 2024; 42:1548-1558. [PMID: 38191663 PMCID: PMC11471619 DOI: 10.1038/s41587-023-02030-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/13/2023] [Indexed: 01/10/2024]
Abstract
The 23 human zinc finger Asp-His-His-Cys motif-containing (ZDHHC) S-acyltransferases catalyze long-chain S-acylation at cysteine residues across an extensive network of hundreds of proteins important for normal physiology or dysregulated in disease. Here we present a technology to directly map the protein substrates of a specific ZDHHC at the whole-proteome level, in intact cells. Structure-guided engineering of paired ZDHHC 'hole' mutants and 'bumped' chemically tagged fatty acid probes enabled probe transfer to specific protein substrates with excellent selectivity over wild-type ZDHHCs. Chemical-genetic systems were exemplified for five human ZDHHCs (3, 7, 11, 15 and 20) and applied to generate de novo ZDHHC substrate profiles, identifying >300 substrates and S-acylation sites for new functionally diverse proteins across multiple cell lines. We expect that this platform will elucidate S-acylation biology for a wide range of models and organisms.
Collapse
Affiliation(s)
| | - Marc P Baggelaar
- The Francis Crick Institute, London, UK
- Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, UK
- Utrecht University, Biomolecular Mass Spectrometry & Proteomics Group, Utrecht, The Netherlands
| | - James Sipthorp
- The Francis Crick Institute, London, UK
- Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, UK
| | - Ana Losada de la Lastra
- The Francis Crick Institute, London, UK
- Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, UK
| | - Manuel Tavares
- The Francis Crick Institute, London, UK
- Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, UK
| | - Jana Volarić
- Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, UK
| | | | - Elisabeth M Storck
- King's College London, Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and Department of Chemistry, London, UK
| | | | - Susana A Palma-Duran
- The Francis Crick Institute, London, UK
- Department of Food Science, Research Center in Food and Development A.C., Hermosillo, Mexico
| | | | | | | | | | - Ulrike S Eggert
- King's College London, Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and Department of Chemistry, London, UK
| | - Edward W Tate
- The Francis Crick Institute, London, UK.
- Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, UK.
| |
Collapse
|
12
|
Pei S, Piao HL. Exploring Protein S-Palmitoylation: Mechanisms, Detection, and Strategies for Inhibitor Discovery. ACS Chem Biol 2024; 19:1868-1882. [PMID: 39160165 DOI: 10.1021/acschembio.4c00110] [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: 08/21/2024]
Abstract
S-palmitoylation is a reversible and dynamic process that involves the addition of long-chain fatty acids to proteins. This protein modification regulates various aspects of protein function, including subcellular localization, stability, conformation, and biomolecular interactions. The zinc finger DHHC (ZDHHC) domain-containing protein family is the main group of enzymes responsible for catalyzing protein S-palmitoylation, and 23 members have been identified in mammalian cells. Many proteins that undergo S-palmitoylation have been linked to disease pathogenesis and progression, suggesting that the development of effective inhibitors is a promising therapeutic strategy. Reducing the protein S-palmitoylation level can target either the PATs directly or their substrates. However, there are rare clinically effective S-palmitoylation inhibitors. This review aims to provide an overview of the S-palmitoylation field, including the catalytic mechanism of ZDHHC, S-palmitoylation detection methods, and the functional impact of protein S-palmitoylation. Additionally, this review focuses on current strategies for expanding the chemical toolbox to develop novel and effective inhibitors that can reduce the level of S-palmitoylation of the target protein.
Collapse
Affiliation(s)
- Shaojun Pei
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, 110122 Shenyang, China
| |
Collapse
|
13
|
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
|
14
|
Shao Y, Hu J, Li H, Lu K. Regulation of autophagy by protein lipidation. ADVANCED BIOTECHNOLOGY 2024; 2:33. [PMID: 39883197 PMCID: PMC11709147 DOI: 10.1007/s44307-024-00040-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/31/2025]
Abstract
Autophagy is a conserved catabolic recycling pathway that can eliminate cytosolic materials to maintain homeostasis and organelle functions. Many studies over the past few decades have demonstrated that abnormal autophagy is associated with a variety of diseases. Protein lipidation plays an important role in the regulation of autophagy by affecting protein trafficking, localization, stability, interactions and signal transduction. Here, we review recent advances in the understanding of the role of lipidation in autophagy, including S-palmitoylation, N-myristoylation, S-prenylation, glycosylphosphatidylinositol (GPI) anchor modification and cholesterylation. We comprehensively review the enzymes and catalytic mechanisms of lipidation and discuss the relationship between lipidation and autophagy, aiming to deepen the understanding of lipidation and promote the discovery of drug targets for the treatment of autophagy-related diseases.
Collapse
Affiliation(s)
- Yuqian Shao
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Junchao Hu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huihui Li
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Kefeng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
15
|
Zhu J, Cao X, Chen Z, Lai B, Xi L, Zhang J, Zhu S, Qi S, Liang Y, Cao F, Zhou B, Song Y, Jiang S, Wang T, Kang X, Kong E. Inhibiting S-palmitoylation arrests metastasis by relocating Rap2b from plasma membrane in colorectal cancer. Cell Death Dis 2024; 15:675. [PMID: 39277583 PMCID: PMC11401852 DOI: 10.1038/s41419-024-07061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Rap2b, a proto-oncogene upregulated in colorectal cancer (CRC), undergoes protein S-palmitoylation at specific C-terminus sites (C176/C177). These palmitoylation sites are crucial for Rap2b localization on the plasma membrane (PM), as mutation of C176 or C177 results in cytosolic relocation of Rap2b. Our study demonstrates that Rap2b influences cell migration and invasion in CRC cells, independent of proliferation, and this activity relies on its palmitoylation. We identify ABHD17a as the depalmitoylating enzyme for Rap2b, altering PM localization and inhibiting cell migration and invasion. EGFR/PI3K signaling regulates Rap2b palmitoylation, with PI3K phosphorylating ABHD17a to modulate its activity. These findings highlight the potential of targeting Rap2b palmitoylation as an intervention strategy. Blocking the C176/C177 sites using an interacting peptide attenuates Rap2b palmitoylation, disrupting PM localization, and suppressing CRC metastasis. This study offers insights into therapeutic approaches targeting Rap2b palmitoylation for the treatment of metastatic CRC, presenting opportunities to improve patient outcomes.
Collapse
Affiliation(s)
- Jiangli Zhu
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Xize Cao
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Zhenshuai Chen
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
- Lankao County Central Hospital, Lankao, China
| | - Birou Lai
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Lingling Xi
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Jinghang Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Xinxiang Medical University, Xinxiang, China
| | - Shaohui Zhu
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shiqian Qi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, China
| | - Yinming Liang
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Fei Cao
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Binhui Zhou
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Yu Song
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Xiaohong Kang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
| | - Eryan Kong
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China.
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
16
|
Forrester MT, Egol JR, Tata A, Tata PR, Foster MW. Analysis of Protein Cysteine Acylation Using a Modified Suspension Trap (Acyl-Trap). J Proteome Res 2024; 23:3716-3725. [PMID: 39008777 PMCID: PMC11826151 DOI: 10.1021/acs.jproteome.4c00225] [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: 07/17/2024]
Abstract
Proteins undergo reversible S-acylation via a thioester linkage in vivo. S-palmitoylation, modification by C16:0 fatty acid, is a common S-acylation that mediates critical protein-membrane and protein-protein interactions. The most widely used S-acylation assays, including acyl-biotin exchange and acyl resin-assisted capture, utilize blocking of free Cys thiols, hydroxylamine-dependent cleavage of the thioester and subsequent labeling of nascent thiol. These assays generally require >500 μg of protein input material per sample and numerous reagent removal and washing steps, making them laborious and ill-suited for high throughput and low input applications. To overcome these limitations, we devised "Acyl-Trap", a suspension trap-based assay that utilizes a thiol-reactive quartz to enable buffer exchange and hydroxylamine-mediated S-acyl enrichment. We show that the method is compatible with protein-level detection of S-acylated proteins (e.g., H-Ras) as well as S-acyl site identification and quantification using "on trap" isobaric labeling and LC-MS/MS from as little as 20 μg of protein input. In mouse brain, Acyl-Trap identified 279 reported sites of S-acylation and 1298 previously unreported putative sites. Also described are conditions for long-term hydroxylamine storage, which streamline the assay. More generally, Acyl-Trap serves as a proof-of-concept for PTM-tailored suspension traps suitable for both traditional protein detection and chemoproteomic workflows.
Collapse
Affiliation(s)
- Michael T. Forrester
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jacob R. Egol
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Purushothama Rao Tata
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Regeneration Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Matthew W. Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
17
|
Ying J, Yang Y, Zhang X, Dong Z, Chen B. Stearoylation cycle regulates the cell surface distribution of the PCP protein Vangl2. Proc Natl Acad Sci U S A 2024; 121:e2400569121. [PMID: 38985771 PMCID: PMC11260150 DOI: 10.1073/pnas.2400569121] [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: 01/10/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
Defects in planar cell polarity (PCP) have been implicated in diverse human pathologies. Vangl2 is one of the core PCP components crucial for PCP signaling. Dysregulation of Vangl2 has been associated with severe neural tube defects and cancers. However, how Vangl2 protein is regulated at the posttranslational level has not been well understood. Using chemical reporters of fatty acylation and biochemical validation, here we present that Vangl2 subcellular localization is regulated by a reversible S-stearoylation cycle. The dynamic process is mainly regulated by acyltransferase ZDHHC9 and deacylase acyl-protein thioesterase 1 (APT1). The stearoylation-deficient mutant of Vangl2 shows decreased plasma membrane localization, resulting in disruption of PCP establishment during cell migration. Genetically or pharmacologically inhibiting ZDHHC9 phenocopies the effects of the stearoylation loss of Vangl2. In addition, loss of Vangl2 stearoylation enhances the activation of oncogenic Yes-associated protein 1 (YAP), serine-threonine kinase AKT, and extracellular signal-regulated protein kinase (ERK) signaling and promotes breast cancer cell growth and HRas G12V mutant (HRasV12)-induced oncogenic transformation. Our results reveal a regulation mechanism of Vangl2, and provide mechanistic insight into how fatty acid metabolism and protein fatty acylation regulate PCP signaling and tumorigenesis by core PCP protein lipidation.
Collapse
Affiliation(s)
- Jiafu Ying
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang321000, China
| | - Yinghong Yang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang321000, China
| | - Xuanpu Zhang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang321000, China
| | - Ze Dong
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang310024, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Baoen Chen
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang321000, China
- Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang311215, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang310000, China
| |
Collapse
|
18
|
Forrester MT, Egol JR, Tata A, Tata PR, Foster MW. Analysis of Protein Cysteine Acylation Using a Modified Suspension Trap (Acyl-Trap). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.23.586403. [PMID: 38585928 PMCID: PMC10996552 DOI: 10.1101/2024.03.23.586403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Proteins undergo reversible S-acylation via a thioester linkage in vivo. S-palmitoylation, modification by C16:0 fatty acid, is a common S-acylation that mediates critical protein-membrane and protein-protein interactions. The most widely used S-acylation assays, including acyl-biotin exchange and acyl resin-assisted capture, utilize blocking of free Cys thiols, hydroxylamine-dependent cleavage of the thioester and subsequent labeling of nascent thiol. These assays generally require >500 micrograms of protein input material per sample and numerous reagent removal and washing steps, making them laborious and ill-suited for high throughput and low input applications. To overcome these limitations, we devised "Acyl-Trap", a suspension trap-based assay that utilizes a thiol-reactive quartz to enable buffer exchange and hydroxylamine-mediated S-acyl enrichment. We show that the method is compatible with protein-level detection of S-acylated proteins (e.g. H-Ras) as well as S-acyl site identification and quantification using "on trap" isobaric labeling and LC-MS/MS from as little as 20 micrograms of protein input. In mouse brain, Acyl-Trap identified 279 reported sites of S-acylation and 1298 previously unreported putative sites. Also described are conditions for long-term hydroxylamine storage, which streamlines the assay. More generally, Acyl-Trap serves as a proof-of-concept for PTM-tailored suspension traps suitable for both traditional protein detection and chemoproteomic workflows.
Collapse
Affiliation(s)
- Michael T. Forrester
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jacob R. Egol
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Purushothama Rao Tata
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Regeneration Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Matthew W. Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
19
|
Choi M, Lee J, Jeong K, Pak Y. Caveolin-2 palmitoylation turnover facilitates insulin receptor substrate-1-directed lipid metabolism by insulin receptor tyrosine kinase. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167173. [PMID: 38631410 DOI: 10.1016/j.bbadis.2024.167173] [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: 10/19/2023] [Revised: 03/13/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
Here, we show that insulin induces palmitoylation turnover of Caveolin-2 (Cav-2) in adipocytes. Acyl protein thioesterases-1 (APT1) catalyzes Cav-2 depalmitoylation, and zinc finger DHHC domain-containing protein palmitoyltransferase 21 (ZDHHC21) repalmitoylation of the depalmitoylated Cav-2 for the turnover, thereby controlling insulin receptor (IR)-Cav-2-insulin receptor substrate-1 (IRS-1)-Akt-driven signaling. Insulin-induced palmitoylation turnover of Cav-2 facilitated glucose uptake and fat storage through induction of lipogenic genes. Cav-2-, APT1-, and ZDHHC21-deficient adipocytes, however, showed increased induction of lipolytic genes and glycerol release. In addition, white adipose tissues from insulin sensitive and resistant obese patients exhibited augmented expression of LYPLA1 (APT1) and ZDHHC20 (ZDHHC20). Our study identifies the specific enzymes regulating Cav-2 palmitoylation turnover, and reveals a new mechanism by which insulin-mediated lipid metabolism is controlled in adipocytes.
Collapse
Affiliation(s)
- Moonjeong Choi
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jaewoong Lee
- Department of Anatomy and Convergence Medical Science, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Kyuho Jeong
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| | - Yunbae Pak
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 52828, Republic of Korea.
| |
Collapse
|
20
|
Liu D, Cruz-Cosme R, Wu Y, Leibowitz J, Tang Q. 2-Bromopalmitate depletes lipid droplets to inhibit viral replication. J Virol 2024; 98:e0017124. [PMID: 38488361 PMCID: PMC11019840 DOI: 10.1128/jvi.00171-24] [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: 01/24/2024] [Accepted: 02/26/2024] [Indexed: 04/17/2024] Open
Abstract
The global impact of emerging viral infections emphasizes the urgent need for effective broad-spectrum antivirals. The cellular organelle, lipid droplet (LD), is utilized by many types of viruses for replication, but its reduction does not affect cell survival. Therefore, LD is a potential target for developing broad-spectrum antivirals. In this study, we found that 2-bromopalmitate (2 BP), a previously defined palmitoylation inhibitor, depletes LD across all studied cell lines and exerts remarkable antiviral effects on different coronaviruses. We comprehensively utilized 2 BP, alongside other palmitoylation inhibitors such as cerulenin and 2-fluoro palmitic acid (2-FPA), as well as the enhancer palmostatin B and evaluated their impact on LD and the replication of human coronaviruses (hCoV-229E, hCoV-Oc43) and murine hepatitis virus (MHV-A59) at non-cytotoxic concentrations. While cerulenin and 2-FPA exhibited moderate inhibition of viral replication, 2 BP exhibited a much stronger suppressive effect on MHV-A59 replication, although they share similar inhibitory effects on palmitoylation. As expected, palmostatin B significantly enhanced viral replication, it failed to rescue the inhibitory effects of 2 BP, whereas it effectively counteracted the effects of cerulenin and 2-FPA. This suggests that the mechanism that 2 BP used to inhibit viral replication is beyond palmitoylation inhibition. Further investigations unveil that 2 BP uniquely depletes LDs, a phenomenon not exhibited by 2-FPA and cerulenin. Importantly, the depletion of LDs was closely associated with the inhibition of viral replication because the addition of oleic acid to 2 BP significantly rescued LD depletion and its inhibitory effects on MHV-A59. Our findings indicate that the inhibitory effects of 2 BP on viral replication primarily stem from LD disruption rather than palmitoylation inhibition. Intriguingly, fatty acid (FA) assays demonstrated that 2 BP reduces the FA level in mitochondria while concurrently increasing FA levels in the cytoplasm. These results highlight the crucial role of LDs in viral replication and uncover a novel biological activity of 2 BP. These insights contribute to the development of broad-spectrum antiviral strategies. IMPORTANCE In our study, we conducted a comparative investigation into the antiviral effects of palmitoylation inhibitors including 2-bromopalmitate (2-BP), 2-fluoro palmitic acid (2-FPA), and cerulenin. Surprisingly, we discovered that 2-BP has superior inhibitory effects on viral replication compared to 2-FPA and cerulenin. However, their inhibitory effects on palmitoylation were the same. Intrigued by this finding, we delved deeper into the underlying mechanism of 2-BP's potent antiviral activity, and we unveiled a novel biological activity of 2-BP: depletion of lipid droplets (LDs). Importantly, we also highlighted the crucial role of LDs in viral replication. Our insights shed new light on the antiviral mechanism of LD depletion paving the way for the development of broad-spectrum antiviral strategies by targeting LDs.
Collapse
Affiliation(s)
- Dongxiao Liu
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | - Julian Leibowitz
- Microbial Pathogenesis and Immunology, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
21
|
Ashford F, Kuo CW, Dunning E, Brown E, Calagan S, Jayasinghe I, Henderson C, Fuller W, Wypijewski K. Cysteine post-translational modifications regulate protein interactions of caveolin-3. FASEB J 2024; 38:e23535. [PMID: 38466300 DOI: 10.1096/fj.202201497rr] [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: 09/15/2022] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
Caveolae are small flask-shaped invaginations of the surface membrane which are proposed to recruit and co-localize signaling molecules. The distinctive caveolar shape is achieved by the oligomeric structural protein caveolin, of which three isoforms exist. Aside from the finding that caveolin-3 is specifically expressed in muscle, functional differences between the caveolin isoforms have not been rigorously investigated. Caveolin-3 is relatively cysteine-rich compared to caveolins 1 and 2, so we investigated its cysteine post-translational modifications. We find that caveolin-3 is palmitoylated at 6 cysteines and becomes glutathiolated following redox stress. We map the caveolin-3 palmitoylation sites to a cluster of cysteines in its C terminal membrane domain, and the glutathiolation site to an N terminal cysteine close to the region of caveolin-3 proposed to engage in protein interactions. Glutathiolation abolishes caveolin-3 interaction with heterotrimeric G protein alpha subunits. Our results indicate that a caveolin-3 oligomer contains up to 66 palmitates, compared to up to 33 for caveolin-1. The additional palmitoylation sites in caveolin-3 therefore provide a mechanistic basis by which caveolae in smooth and striated muscle can possess unique phospholipid and protein cargoes. These unique adaptations of the muscle-specific caveolin isoform have important implications for caveolar assembly and signaling.
Collapse
Affiliation(s)
- Fiona Ashford
- School of Medicine, University of Dundee, Dundee, UK
| | - Chien-Wen Kuo
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Emma Dunning
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Elaine Brown
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Sarah Calagan
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Izzy Jayasinghe
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | | | - William Fuller
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Krzysztof Wypijewski
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
- School of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
22
|
Wang Y, Shen N, Yang Y, Xia Y, Zhang W, Lu Y, Wang Z, Yang Z, Wang Z. ZDHHC5-mediated S-palmitoylation of FAK promotes its membrane localization and epithelial-mesenchymal transition in glioma. Cell Commun Signal 2024; 22:46. [PMID: 38233791 PMCID: PMC10795333 DOI: 10.1186/s12964-023-01366-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: 08/05/2023] [Accepted: 10/26/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Abnormal activation of FAK is associated with tumor development and metastasis. Through interactions with other intracellular signalling molecules, FAK influences cytoskeletal remodelling, modulation of adhesion signalling, and activation of transcription factors, promoting migration and invasion of tumor cells. However, the exact mechanism that regulates these processes remains unresolved. Herein, our findings indicate that the S-palmitoylation of FAK is crucial for both its membrane localization and activation. METHODS The palmitoylation of FAK in U251 and T98G cells was assessed by an acyl-PEG exchange (APE) assay and a metabolic incorporation assay. Cellular palmitoylation was inhibited using 2-bromopalmitate, and the palmitoylation status and cellular localization of FAK were determined. A metabolic incorporation assay was used to identify the potential palmitoyl acyltransferase and the palmitoylation site of FAK. Cell Counting Kit-8 (CCK8) assays, colony formation assays, and Transwell assays were conducted to assess the impact of ZDHHC5 in GBM. Additionally, intracranial GBM xenografts were utilized to investigate the effects of genetically silencing ZDHHC5 on tumor growth. RESULTS Inhibiting FAK palmitoylation leads to its redistribution from the membrane to the cytoplasm and a decrease in its phosphorylation. Moreover, ZDHHC5, a protein-acyl-transferase (PAT), catalyzes this key modification of FAK at C456. Knockdown of ZDHHC5 abrogates the S-palmitoylation and membrane distribution of FAK and impairs cell proliferation, invasion, and epithelial-mesenchymal transition (EMT). Taken together, our research reveals the crucial role of ZDHHC5 as a PAT responsible for FAK S-palmitoylation, membrane localization, and activation. CONCLUSIONS These results imply that targeting the ZDHHC5/FAK axis has the potential to be a promising strategy for therapeutic interventions for glioblastoma (GBM). Video Abstract.
Collapse
Affiliation(s)
- Yang Wang
- Center for Clinical Medical Research, the Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Na Shen
- Center for Clinical Medical Research, the Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yang Yang
- Department of Pediatric Surgery, the Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yuan Xia
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wenhao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yu Lu
- Department of Orthopedics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233099, China
| | - Zhicheng Wang
- Department of Orthopedics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233099, China
| | - Ze Yang
- Department of Pediatric Surgery, the Affiliated Hospital of Nantong University, Nantong, 226001, China.
| | - Zhangjie Wang
- Center for Clinical Medical Research, the Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Das T, Hang HC. Discovery and Characterization of IFITM S-Palmitoylation. Viruses 2023; 15:2329. [PMID: 38140570 PMCID: PMC10747768 DOI: 10.3390/v15122329] [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: 11/01/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Interferon-induced transmembrane proteins (IFITM1, 2 and 3) are important host antiviral defense factors. They are active against viruses like the influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV) and severe acute respiratory syndrome coronavirus (SARS-CoV). In this review, we focus on IFITM3 S-palmitoylation, a reversible lipid modification, and describe its role in modulating IFITM3 antiviral activity. Our laboratory discovered S-palmitoylation of IFITMs using chemical proteomics and demonstrated the importance of highly conserved fatty acid-modified Cys residues in IFITM3 antiviral activity. Further studies showed that site-specific S-palmitoylation at Cys72 is important for IFITM3 trafficking to restricted viruses (IAV and EBOV) and membrane-sterol interactions. Thus, site-specific lipid modification of IFITM3 directly regulates its antiviral activity, cellular trafficking, and membrane-lipid interactions.
Collapse
Affiliation(s)
- Tandrila Das
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Howard C. Hang
- Departments of Immunology and Microbiology and Chemistry, Scripps Research, La Jolla, CA 92037, USA
| |
Collapse
|
26
|
Gu M, Jiang H, Tan M, Yu L, Xu N, Li Y, Wu H, Hou Q, Dai C. Palmitoyltransferase DHHC9 and acyl protein thioesterase APT1 modulate renal fibrosis through regulating β-catenin palmitoylation. Nat Commun 2023; 14:6682. [PMID: 37865665 PMCID: PMC10590414 DOI: 10.1038/s41467-023-42476-z] [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: 02/08/2023] [Accepted: 10/12/2023] [Indexed: 10/23/2023] Open
Abstract
palmitoylation, a reversible post-translational modification, is initiated by the DHHC family of palmitoyltransferases and reversed by several acyl protein thioesterases. However, the role and mechanisms for protein palmitoylation in renal fibrosis have not been elucidated. Here we show protein palmitoylation and DHHC9 were downregulated in the fibrotic kidneys of mouse models and chronic kidney disease (CKD) patients. Ablating DHHC9 in tubular cells aggravated, while inducing DHHC9 overexpression with adeno-DHHC9 transfection or iproniazid treatment protected against kidney fibrosis in male mouse models. Mechanistically, DHHC9 palmitoylated β-catenin, thereby promoted its ubiquitination and degradation. Additionally, acyl protein thioesterase 1 (APT1) was induced in the fibrotic kidneys, which depalmitoylated β-catenin, increased its abundance and nuclear translocation. Ablating tubular APT1 or inhibiting APT1 with ML348 markedly protected against unilateral ureter obstruction (UUO) or ischemia/reperfusion injury (IRI)-induced kidney fibrosis in male mice. This study reveals the regulatory mechanism of protein palmitoylation in kidney fibrosis.
Collapse
Affiliation(s)
- Mengru Gu
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
- Department of Clinical Genetics, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Hanlu Jiang
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Mengzhu Tan
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Long Yu
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Ning Xu
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Ying Li
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Han Wu
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Qing Hou
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Chunsun Dai
- Center for Kidney Diseases, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China.
- Department of Clinical Genetics, the Second Affiliated Hospital of Nanjing Medical University; Nanjing, China, 210009, 262 North Zhongshan Road, Nanjing, Jiangsu, China.
| |
Collapse
|
27
|
Yucel BP, Al Momany EM, Evans AJ, Seager R, Wilkinson KA, Henley JM. Coordinated interplay between palmitoylation, phosphorylation and SUMOylation regulates kainate receptor surface expression. Front Mol Neurosci 2023; 16:1270849. [PMID: 37868810 PMCID: PMC10585046 DOI: 10.3389/fnmol.2023.1270849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Kainate receptors (KARs) are key regulators of neuronal excitability and synaptic transmission. KAR surface expression is tightly controlled in part by post-translational modifications (PTMs) of the GluK2 subunit. We have shown previously that agonist activation of GluK2-containing KARs leads to phosphorylation of GluK2 at S868, which promotes subsequent SUMOylation at K886 and receptor endocytosis. Furthermore, GluK2 has been shown to be palmitoylated. However, how the interplay between palmitoylation, phosphorylation and SUMOylation orchestrate KAR trafficking remains unclear. Here, we used a library of site-specific GluK2 mutants to investigate the interrelationship between GluK2 PTMs, and their impact on KAR surface expression. We show that GluK2 is basally palmitoylated and that this is decreased by kainate (KA) stimulation. Moreover, a non-palmitoylatable GluK2 mutant (C858/C871A) shows enhanced S868 phosphorylation and K886 SUMOylation under basal conditions and is insensitive to KA-induced internalisation. These results indicate that GluK2 palmitoylation contributes to stabilising KAR surface expression and that dynamic depalmitoylation promotes downstream phosphorylation and SUMOylation to mediate activity-dependent KAR endocytosis.
Collapse
Affiliation(s)
| | | | | | | | - Kevin A. Wilkinson
- Centre for Synaptic Plasticity, School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Jeremy M. Henley
- Centre for Synaptic Plasticity, School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
28
|
Li M, Zhang L, Chen CW. Diverse Roles of Protein Palmitoylation in Cancer Progression, Immunity, Stemness, and Beyond. Cells 2023; 12:2209. [PMID: 37759431 PMCID: PMC10526800 DOI: 10.3390/cells12182209] [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: 07/20/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Protein S-palmitoylation, a type of post-translational modification, refers to the reversible process of attachment of a fatty acyl chain-a 16-carbon palmitate acid-to the specific cysteine residues on target proteins. By adding the lipid chain to proteins, it increases the hydrophobicity of proteins and modulates protein stability, interaction with effector proteins, subcellular localization, and membrane trafficking. Palmitoylation is catalyzed by a group of zinc finger DHHC-containing proteins (ZDHHCs), whereas depalmitoylation is catalyzed by a family of acyl-protein thioesterases. Increasing numbers of oncoproteins and tumor suppressors have been identified to be palmitoylated, and palmitoylation is essential for their functions. Understanding how palmitoylation influences the function of individual proteins, the physiological roles of palmitoylation, and how dysregulated palmitoylation leads to pathological consequences are important drivers of current research in this research field. Further, due to the critical roles in modifying functions of oncoproteins and tumor suppressors, targeting palmitoylation has been used as a candidate therapeutic strategy for cancer treatment. Here, based on recent literatures, we discuss the progress of investigating roles of palmitoylation in regulating cancer progression, immune responses against cancer, and cancer stem cell properties.
Collapse
Affiliation(s)
- Mingli Li
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
| | - Leisi Zhang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
29
|
Nůsková H, Cortizo FG, Schwenker LS, Sachsenheimer T, Diakonov EE, Tiebe M, Schneider M, Lohbeck J, Reid C, Kopp-Schneider A, Helm D, Brügger B, Miller AK, Teleman AA. Competition for cysteine acylation by C16:0 and C18:0 derived lipids is a global phenomenon in the proteome. J Biol Chem 2023; 299:105088. [PMID: 37495107 PMCID: PMC10470219 DOI: 10.1016/j.jbc.2023.105088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023] Open
Abstract
S-acylation is a reversible posttranslational protein modification consisting of attachment of a fatty acid to a cysteine via a thioester bond. Research over the last few years has shown that a variety of different fatty acids, such as palmitic acid (C16:0), stearate (C18:0), or oleate (C18:1), are used in cells to S-acylate proteins. We recently showed that GNAI proteins can be acylated on a single residue, Cys3, with either C16:0 or C18:1, and that the relative proportion of acylation with these fatty acids depends on the level of the respective fatty acid in the cell's environment. This has functional consequences for GNAI proteins, with the identity of the acylating fatty acid affecting the subcellular localization of GNAIs. Unclear is whether this competitive acylation is specific to GNAI proteins or a more general phenomenon in the proteome. We perform here a proteome screen to identify proteins acylated with different fatty acids. We identify 218 proteins acylated with C16:0 and 308 proteins acylated with C18-lipids, thereby uncovering novel targets of acylation. We find that most proteins that can be acylated by C16:0 can also be acylated with C18-fatty acids. For proteins with more than one acylation site, we find that this competitive acylation occurs on each individual cysteine residue. This raises the possibility that the function of many different proteins can be regulated by the lipid environment via differential S-acylation.
Collapse
Affiliation(s)
- Hana Nůsková
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabiola Garcia Cortizo
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Sophie Schwenker
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Egor E Diakonov
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcel Tiebe
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Schneider
- Mass Spectrometry Based Protein Analysis Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jasmin Lohbeck
- Research Group Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carissa Reid
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Dominic Helm
- Mass Spectrometry Based Protein Analysis Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Aubry K Miller
- Research Group Cancer Drug Development, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aurelio A Teleman
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
30
|
Huang J, Yang JG, Ren JG, Xia HF, Chen GH, Fu QY, Zhang LZ, Liu HM, Wang KM, Xie QH, Chen G. Overexpression of RAB27A in Oral Squamous Cell Carcinoma Promotes Tumor Migration and Invasion via Modulation of EGFR Membrane Stability. Int J Mol Sci 2023; 24:13103. [PMID: 37685910 PMCID: PMC10488256 DOI: 10.3390/ijms241713103] [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: 07/30/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most prevalent subtype of head and neck tumors, highly prone to lymph node metastasis. This study aims to examine the expression pattern of Ras-related protein Rab-27A (RAB27A) and explore its potential implications in OSCC. The expression of RAB27A was assessed through immunohistochemical analysis utilizing tissue microarrays. In vitro experiments were conducted using RAB27A-knockdown cells to investigate its impact on OSCC tumor cells. Additionally, transcriptome sequencing was performed to elucidate potential underlying mechanisms. RAB27A was significantly overexpressed in OSCC, and particularly in metastatic lymph nodes. It was positively correlated with the clinical progression and poor survival prognosis. Silencing RAB27A notably decreased the proliferation, migration, and invasion abilities of OSCC cells in vitro. A Gene Ontology (GO) enrichment analysis indicated a strong association between RAB27A and the epidermal growth factor receptor (EGFR) signaling pathway. Further investigations revealed that RAB27A regulated the palmitoylation of EGFR via zinc finger DHHC-type containing 13 (ZDHHC13). These findings provide insights into OSCC progression and highlight RAB27A as a potential therapeutic target for combating this aggressive cancer.
Collapse
Affiliation(s)
- Jue Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Jie-Gang Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jian-Gang Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Hou-Fu Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Gao-Hong Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Qiu-Yun Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Lin-Zhou Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Kui-Ming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Qi-Hui Xie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China (H.-M.L.)
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
31
|
Geddes JW, Bondada V, Croall DE, Rodgers DW, Gal J. Impaired activity and membrane association of most calpain-5 mutants causal for neovascular inflammatory vitreoretinopathy. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166747. [PMID: 37207905 PMCID: PMC10332796 DOI: 10.1016/j.bbadis.2023.166747] [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: 07/22/2022] [Revised: 03/29/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023]
Abstract
Neovascular inflammatory vitreoretinopathy (NIV) is a rare eye disease that ultimately leads to complete blindness and is caused by mutations in the gene encoding calpain-5 (CAPN5), with six pathogenic mutations identified. In transfected SH-SY5Y cells, five of the mutations resulted in decreased membrane association, diminished S-acylation, and reduced calcium-induced autoproteolysis of CAPN5. CAPN5 proteolysis of the autoimmune regulator AIRE was impacted by several NIV mutations. R243, L244, K250 and the adjacent V249 are on β-strands in the protease core 2 domain. Conformational changes induced by Ca2+binding result in these β-strands forming a β-sheet and a hydrophobic pocket which docks W286 side chain away from the catalytic cleft, enabling calpain activation based on comparison with the Ca2+-bound CAPN1 protease core. The pathologic variants R243L, L244P, K250N, and R289W are predicted to disrupt the β-strands, β-sheet, and hydrophobic pocket, impairing calpain activation. The mechanism by which these variants impair membrane association is unclear. G376S impacts a conserved residue in the CBSW domain and is predicted to disrupt a loop containing acidic residues which may contribute to membrane binding. G267S did not impair membrane association and resulted in a slight but significant increase in autoproteolytic and proteolytic activity. However, G267S is also identified in individuals without NIV. Combined with the autosomal dominant pattern of NIV inheritance and evidence that CAPN5 may dimerize, the results are consistent with a dominant negative mechanism for the five pathogenic variants which resulted in impaired CAPN5 activity and membrane association and a gain-of-function for the G267S variant.
Collapse
Affiliation(s)
- James W Geddes
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| | - Vimala Bondada
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA
| | - Dorothy E Croall
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA.
| | - David W Rodgers
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, KY 40536, USA.
| | - Jozsef Gal
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
32
|
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
|
33
|
Zhang Q, Sjögren B. Palmitoylation of RGS20 affects Gα o-mediated signaling independent of its GAP activity. Cell Signal 2023; 107:110682. [PMID: 37075876 DOI: 10.1016/j.cellsig.2023.110682] [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: 09/13/2022] [Revised: 04/07/2023] [Accepted: 04/15/2023] [Indexed: 04/21/2023]
Abstract
Regulator of protein signaling (RGS20) is a member of the RGS protein superfamily, which serve as key negative regulators of G protein-mediated signal transduction. Through their GTPase accelerating protein (GAP) activity, RGS proteins deactivate α-subunits of heterotrimeric G proteins. In addition, the majority of RGS proteins also have the ability to act through other, non-GAP related, functions. RGS20 is one of three members of the RZ subfamily, which all show selective GAP activity towards Gαz, however emerging data suggest that RGS20 can also regulate Gi/o-mediated signaling. While increased RGS20 expression is associated with the progression of multiple cancers, a large gap still exists relating to the mechanisms of RGS20 regulation and function. RGS20 contains a poly-cysteine string motif and a conserved cysteine in RGS domain, which are assumed to be palmitoylated. Palmitoylation, an important post-translational modification, plays an important role in cells by changing cellular functions of proteins. Consequently, the aim of this study was to confirm that RGS20 is palmitoylated and determine how palmitoylation affects its inhibition of Gαo-mediated signaling. We found a significant positive correlation between RGS20 palmitoylation and its association with active Gαo. We also showed that a conserved cysteine residue in the RGS domain is a critical site for its palmitoylation, with large impact on its association with Gαo. Palmitoylation on this site did not affect its GAP activity, however, it increased the inhibition of Gαo-mediated cAMP signaling. Altogether these data suggest that palmitoylation is a regulatory mechanism controlling RGS20 function, and that RGS20 can inhibit Gαo signaling through both GAP activity and non-GAP mechanisms.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States of America
| | - Benita Sjögren
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States of America.
| |
Collapse
|
34
|
Kuo CWS, Dobi S, Gök C, Da Silva Costa A, Main A, Robertson-Gray O, Baptista-Hon D, Wypijewski KJ, Costello H, Hales TG, MacQuaide N, Smith GL, Fuller W. Palmitoylation of the pore-forming subunit of Ca(v)1.2 controls channel voltage sensitivity and calcium transients in cardiac myocytes. Proc Natl Acad Sci U S A 2023; 120:e2207887120. [PMID: 36745790 PMCID: PMC9963536 DOI: 10.1073/pnas.2207887120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 12/07/2022] [Indexed: 02/08/2023] Open
Abstract
Mammalian voltage-activated L-type Ca2+ channels, such as Ca(v)1.2, control transmembrane Ca2+ fluxes in numerous excitable tissues. Here, we report that the pore-forming α1C subunit of Ca(v)1.2 is reversibly palmitoylated in rat, rabbit, and human ventricular myocytes. We map the palmitoylation sites to two regions of the channel: The N terminus and the linker between domains I and II. Whole-cell voltage clamping revealed a rightward shift of the Ca(v)1.2 current-voltage relationship when α1C was not palmitoylated. To examine function, we expressed dihydropyridine-resistant α1C in human induced pluripotent stem cell-derived cardiomyocytes and measured Ca2+ transients in the presence of nifedipine to block the endogenous channels. The transients generated by unpalmitoylatable channels displayed a similar activation time course but significantly reduced amplitude compared to those generated by wild-type channels. We thus conclude that palmitoylation controls the voltage sensitivity of Ca(v)1.2. Given that the identified Ca(v)1.2 palmitoylation sites are also conserved in most Ca(v)1 isoforms, we propose that palmitoylation of the pore-forming α1C subunit provides a means to regulate the voltage sensitivity of voltage-activated Ca2+ channels in excitable cells.
Collapse
Affiliation(s)
- Chien-Wen S. Kuo
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Sara Dobi
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Caglar Gök
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Ana Da Silva Costa
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Alice Main
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Olivia Robertson-Gray
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Daniel Baptista-Hon
- Division of Systems Medicine, Institute of Academic Anaesthesia, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078China
| | | | - Hannah Costello
- Division of Systems Medicine, Institute of Academic Anaesthesia, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Tim G. Hales
- Division of Systems Medicine, Institute of Academic Anaesthesia, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Niall MacQuaide
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - Godfrey L. Smith
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| | - William Fuller
- School of Cardiovascular and Metabolic Health, University of Glasgow, GlasgowG12 8QQ, UK
| |
Collapse
|
35
|
Ravishankar R, Hildebrandt ER, Greenway G, Asad N, Gore S, Dore TM, Schmidt WK. Specific Disruption of Ras2 CAAX Proteolysis Alters Its Localization and Function. Microbiol Spectr 2023; 11:e0269222. [PMID: 36602340 PMCID: PMC9927470 DOI: 10.1128/spectrum.02692-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Many CAAX proteins, such as Ras GTPase, undergo a series of posttranslational modifications at their carboxyl terminus (i.e., cysteine prenylation, endoproteolysis of AAX, and carboxylmethylation). Some CAAX proteins, however, undergo prenylation-only modification, such as Saccharomyces cerevisiae Hsp40 Ydj1. We previously observed that altering the CAAX motif of Ydj1 from prenylation-only to canonical resulted in altered Ydj1 function and localization. Here, we investigated the effects of a reciprocal change that altered the well-characterized canonical CAAX motif of S. cerevisiae Ras2 to prenylation-only. We observed that the type of CAAX motif impacted Ras2 protein levels, localization, and function. Moreover, we observed that using a prenylation-only sequence to stage hyperactive Ras2-G19V as a farnesylated and nonproteolyzed intermediate resulted in a different phenotype relative to staging by a genetic RCE1 deletion strategy that simultaneously affected many CAAX proteins. These findings suggested that a prenylation-only CAAX motif is useful for probing the specific impact of CAAX proteolysis on Ras2 under conditions where other CAAX proteins are normally modified. We propose that our strategy could be easily applied to a wide range of CAAX proteins for examining the specific impact of CAAX proteolysis on their functions. IMPORTANCE CAAX proteins are subject to multiple posttranslational modifications: cysteine prenylation, CAAX proteolysis, and carboxylmethylation. For investigations of CAAX proteolysis, this study took the novel approach of using a proteolysis-resistant CAAX sequence to stage Saccharomyces cerevisiae Ras2 GTPase in a farnesylated and nonproteolyzed state. Our approach specifically limited the effects of disrupting CAAX proteolysis to Ras2. This represented an improvement over previous methods where CAAX proteolysis was inhibited by gene knockout, small interfering RNA knockdown, or biochemical inhibition of the Rce1 CAAX protease, which can lead to pleiotropic and unclear attribution of effects due to the action of Rce1 on multiple CAAX proteins. Our approach yielded results that demonstrated specific impacts of CAAX proteolysis on the function, localization, and other properties of Ras2, highlighting the utility of this approach for investigating the impact of CAAX proteolysis in other protein contexts.
Collapse
Affiliation(s)
- Rajani Ravishankar
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Emily R. Hildebrandt
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Grace Greenway
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Nadeem Asad
- New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Sangram Gore
- New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Timothy M. Dore
- New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| | - Walter K. Schmidt
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
36
|
Jiménez-Munguía I, Beaven AH, Blank PS, Sodt AJ, Zimmerberg J. Interferon-induced transmembrane protein 3 (IFITM3) and its antiviral activity. Curr Opin Struct Biol 2022; 77:102467. [PMID: 36306674 DOI: 10.1016/j.sbi.2022.102467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 01/30/2023]
Abstract
Infections caused by enveloped viruses require fusion with cellular membranes for viral genome entry. Viral entry occurs following an interaction of viral and cellular membranes allowing the formation of fusion pores, by which the virus accesses the cytoplasm. Here, we focus on interferon-induced transmembrane protein 3 (IFITM3) and its antiviral activity. IFITM3 is predicted to block or stall viral fusion at an intermediate state, causing viral propagation to fail. After introducing IFITM3, we describe the generalized lipid membrane fusion pathway and how it can be stalled, particularly with respect to IFITM3, and current questions regarding IFITM3's topology, with specific emphasis on IFITM3's amphipathic α-helix (AAH) 59V-68M, which is necessary for the antiviral activity. We report new hydrophobicity and hydrophobic moment calculations for this peptide and a variety of active site peptides from known membrane-remodeling proteins. Finally, we discuss the effects of posttranslational modifications and localization, how IFITM3's AAH may block viral fusion, and possible ramifications of membrane composition.
Collapse
Affiliation(s)
- I Jiménez-Munguía
- Section on Integrative Biophysics Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), MD, USA
| | - A H Beaven
- Unit on Membrane Chemical Physics Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH) MD, USA; Postdoctoral Research Associate Program, National Institute of General Medical Sciences National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - P S Blank
- Section on Integrative Biophysics Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), MD, USA
| | - A J Sodt
- Unit on Membrane Chemical Physics Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH) MD, USA.
| | - J Zimmerberg
- Section on Integrative Biophysics Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), MD, USA.
| |
Collapse
|
37
|
Zheng S, Lin J, Pang Z, Zhang H, Wang Y, Ma L, Zhang H, Zhang X, Chen M, Zhang X, Zhao C, Qi J, Cao L, Wang M, He X, Sheng R. Aberrant Cholesterol Metabolism and Wnt/β-Catenin Signaling Coalesce via Frizzled5 in Supporting Cancer Growth. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200750. [PMID: 35975457 PMCID: PMC9534957 DOI: 10.1002/advs.202200750] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/23/2022] [Indexed: 05/12/2023]
Abstract
Frizzled (Fzd) proteins are Wnt receptors and play essential roles in development, homeostasis, and oncogenesis. How Wnt/Fzd signaling is coupled to physiological regulation remains unknown. Cholesterol is reported as a signaling molecule regulating morphogen such as Hedgehog signaling. Despite the elusiveness of the in-depth mechanism, it is well-established that pancreatic cancer specially requires abnormal cholesterol metabolism levels for growth. In this study, it is unexpectedly found that among ten Fzds, Fzd5 has a unique capacity to bind cholesterol specifically through its conserved extracellular linker region. Cholesterol-binding enables Fzd5 palmitoylation, which is indispensable for receptor maturation and trafficking to the plasma membrane. In Wnt-addicted pancreatic ductal adenocarcinoma (PDAC), cholesterol stimulates tumor growth via Fzd5-mediated Wnt/β-catenin signaling. A natural oxysterol, 25-hydroxylsterol competes with cholesterol and inhibits Fzd5 maturation and Wnt signaling, thereby alleviating PDAC growth. This cholesterol-receptor interaction and ensuing receptor lipidation uncover a novel mechanism by which Fzd5 acts as a cholesterol sensor and pivotal connection coupling lipid metabolism to morphogen signaling. These findings further suggest that cholesterol-targeting may provide new therapeutic opportunities for treating Wnt-dependent cancers.
Collapse
Affiliation(s)
- Shaoqin Zheng
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Jiahui Lin
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Zhongqiu Pang
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Hui Zhang
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Yinuo Wang
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Lanjing Ma
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Haijiao Zhang
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
| | - Xi Zhang
- College of SciencesNortheastern UniversityShenyang110004P. R. China
| | - Maorong Chen
- F.M Kirby Neurobiology CenterBoston Children's HospitalDepartment of NeurologyHarvard Medical SchoolBostonMA02115USA
| | - Xinjun Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationNational Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Chao Zhao
- School of Public HealthJilin UniversityChangchun130021P. R. China
| | - Jun Qi
- Department of Cancer BiologyDana‐Farber Cancer InstituteDepartment of MedicineHarvard Medical SchoolBostonMA02215USA
| | - Liu Cao
- Institute of Translational MedicineKey Laboratory of Cell Biology of Ministry of Public Healthand Key Laboratory of Medical Cell Biology of Ministry of EducationLiaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and PreventionChina Medical UniversityShenyang110112P. R. China
| | - Min Wang
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji HospitalTongji Medical CollegeHuazhong University of Science and Technology1095 Jiefang AveWuhan430030P. R. China
| | - Xi He
- F.M Kirby Neurobiology CenterBoston Children's HospitalDepartment of NeurologyHarvard Medical SchoolBostonMA02115USA
| | - Ren Sheng
- College of Life and Health ScienceNortheastern UniversityShenyang110819P. R. China
- F.M Kirby Neurobiology CenterBoston Children's HospitalDepartment of NeurologyHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
38
|
Gal J, Bondada V, Mashburn CB, Rodgers DW, Croall DE, Geddes JW. S-acylation regulates the membrane association and activity of Calpain-5. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119298. [PMID: 35643222 DOI: 10.1016/j.bbamcr.2022.119298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/05/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Calpain-5 (CAPN5) is a member of the calpain family of calcium-activated neutral thiol proteases. CAPN5 is partly membrane associated, despite its lack of a transmembrane domain. Unlike classical calpains, CAPN5 contains a C-terminal C2 domain. C2 domains often have affinity to lipids, mediating membrane association. We recently reported that the C2 domain of CAPN5 was essential for its membrane association and the activation of its autolytic activity. However, despite the removal of the C2 domain by autolysis, the N-terminal fragment of CAPN5 remained membrane associated. S-acylation, also referred to as S-palmitoylation, is a reversible post-translational lipid modification of cysteine residues that promotes membrane association of soluble proteins. In the present study several S-acylated cysteine residues were identified in CAPN5 with the acyl-PEG exchange method. Data reported here demonstrate that CAPN5 is S-acylated on up to three cysteine residues including Cys-4 and Cys-512, and likely Cys-507. The D589N mutation in a potential calcium binding loop within the C2 domain interfered with the S-acylation of CAPN5, likely preventing initial membrane association. Mutating specific cysteine residues of CAPN5 interfered with both its membrane association and the activation of CAPN5 autolysis. Taken together, our results suggest that the S-acylation of CAPN5 is critical for its membrane localization which appears to favor its enzymatic activity.
Collapse
Affiliation(s)
- Jozsef Gal
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| | - Vimala Bondada
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA
| | - Charles B Mashburn
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA
| | - David W Rodgers
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Dorothy E Croall
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA
| | - James W Geddes
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
39
|
Das T, Yang X, Lee H, Garst EH, Valencia E, Chandran K, Im W, Hang HC. S-Palmitoylation and Sterol Interactions Mediate Antiviral Specificity of IFITMs. ACS Chem Biol 2022; 17:2109-2120. [PMID: 35861660 PMCID: PMC10597057 DOI: 10.1021/acschembio.2c00176] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Interferon-induced transmembrane proteins (IFITM1, 2, and 3) are important antiviral proteins that are active against many viruses, including influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV), and severe acute respiratory syndrome coronavirus (SARS-CoV). IFITM proteins exhibit specificity in activity, but their distinct mechanisms of action and regulation are unclear. Since S-palmitoylation and cholesterol homeostasis are crucial for viral infections, we investigated IFITM interactions with cholesterol by photoaffinity cross-linking in mammalian cells along with molecular dynamic simulations and nuclear magnetic resonance analysis in vitro. These studies suggest that cholesterol can directly interact with S-palmitoylated IFITMs in cells and alter the conformation of IFITMs in membrane bilayers. Notably, we discovered that the S-palmitoylation levels regulate differential IFITM protein interactions with cholesterol in mammalian cells and specificity of antiviral activity toward IAV, SARS-CoV-2, and EBOV. Our studies suggest that modulation of IFITM S-palmitoylation levels and cholesterol interaction influence host susceptibility to different viruses.
Collapse
Affiliation(s)
- Tandrila Das
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, NY 10065, United States
- Tri-Institutional Ph.D. Program in Chemical Biology, New York, NY 10065, United States
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
| | - Xinglin Yang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
| | - Hwayoung Lee
- Department of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA 18015, United States
| | - Emma H. Garst
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, NY 10065, United States
- Tri-Institutional Ph.D. Program in Chemical Biology, New York, NY 10065, United States
| | - Estefania Valencia
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Wonpil Im
- Department of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA 18015, United States
| | - Howard C. Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
- Department of Chemistry, Scripps Research, La Jolla, CA 92037, United States
| |
Collapse
|
40
|
Emenike B, Nwajiobi O, Raj M. Covalent Chemical Tools for Profiling Post-Translational Modifications. Front Chem 2022; 10:868773. [PMID: 35860626 PMCID: PMC9289218 DOI: 10.3389/fchem.2022.868773] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/30/2022] [Indexed: 12/05/2022] Open
Abstract
Nature increases the functional diversity of the proteome through posttranslational modifications (PTMs); a process that involves the proteolytic processing or catalytic attachment of diverse functional groups onto proteins. These modifications modulate a host of biological activities and responses. Consequently, anomalous PTMs often correlate to a host of diseases, hence there is a need to detect these transformations, both qualitatively and quantitatively. One technique that has gained traction is the use of robust chemical strategies to label different PTMs. By utilizing the intrinsic chemical reactivity of the different chemical groups on the target amino acid residues, this strategy can facilitate the delineation of the overarching and inclusionary roles of these different modifications. Herein, we will discuss the current state of the art in post-translational modification analysis, with a direct focus on covalent chemical methods used for detecting them.
Collapse
|
41
|
Tien CF, Tsai WT, Chen CH, Chou HJ, Zhang MM, Lin JJ, Lin EJ, Dai SS, Ping YH, Yu CY, Kuo YP, Tsai WH, Chen HW, Yu GY. Glycosylation and S-palmitoylation regulate SARS-CoV-2 spike protein intracellular trafficking. iScience 2022; 25:104709. [PMID: 35813875 PMCID: PMC9250814 DOI: 10.1016/j.isci.2022.104709] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/19/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Post-translational modifications (PTMs), such as glycosylation and palmitoylation, are critical to protein folding, stability, intracellular trafficking, and function. Understanding regulation of PTMs of SARS-CoV-2 spike (S) protein could help the therapeutic drug design. Herein, the VSV vector was used to produce SARS-CoV-2 S pseudoviruses to examine the roles of the 611LYQD614 and cysteine-rich motifs in S protein maturation and virus infectivity. Our results show that 611LY612 mutation alters S protein intracellular trafficking and reduces cell surface expression level. It also changes S protein glycosylation pattern and decreases pseudovirus infectivity. The S protein contains four cysteine-rich clusters with clusters I and II as the main palmitoylation sites. Mutations of clusters I and II disrupt S protein trafficking from ER-to-Golgi, suppress pseudovirus production, and reduce spike-mediated membrane fusion activity. Taken together, glycosylation and palmitoylation orchestrate the S protein maturation processing and are critical for S protein-mediated membrane fusion and infection.
611LY612 mutation alters the glycosylation pattern of the SARS-CoV-2 S protein 611LY612 mutation reduces S protein surface expression level Palmitoylation targets mature S protein to the Golgi and plasma membrane Palmitoylation is required for pseudovirus and SARS-CoV-2 production
Collapse
|
42
|
Mekhail K, Lee M, Sugiyama M, Astori A, St-Germain J, Latreille E, Khosraviani N, Wei K, Li Z, Rini J, Lee WL, Antonescu C, Raught B, Fairn GD. Fatty Acid Synthase inhibitor TVB-3166 prevents S-acylation of the Spike protein of human coronaviruses. J Lipid Res 2022; 63:100256. [PMID: 35921881 PMCID: PMC9339154 DOI: 10.1016/j.jlr.2022.100256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/24/2022] Open
Abstract
The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other coronaviruses mediates host cell entry and is S-acylated on multiple phylogenetically conserved cysteine residues. Multiple protein acyltransferase enzymes have been reported to post-translationally modify spike proteins; however, strategies to exploit this modification are lacking. Using resin-assisted capture MS, we demonstrate that the spike protein is S-acylated in SARS-CoV-2-infected human and monkey epithelial cells. We further show that increased abundance of the acyltransferase ZDHHC5 associates with increased S-acylation of the spike protein, whereas ZDHHC5 knockout cells had a 40% reduction in the incorporation of an alkynyl-palmitate using click chemistry detection. We also found that the S-acylation of the spike protein is not limited to palmitate, as clickable versions of myristate and stearate were also labelled the protein. Yet, we observed that ZDHHC5 was only modified when incubated with alkyne-palmitate, suggesting it has specificity for this acyl-CoA, and that other ZDHHC enzymes may use additional fatty acids to modify the spike protein. Since multiple ZDHHC isoforms may modify the spike protein, we also examined the ability of the FASN inhibitor TVB-3166 to prevent S-acylation of the spike proteins of SARS-CoV-2 and human CoV-229E. We show that treating cells with TVB-3166 inhibited S-acylation of expressed spike proteins and attenuated the ability of SARS-CoV-2 and human CoV-229E to spread in vitro. Our findings further substantiate the necessity of CoV spike protein S-acylation and demonstrate that de novo fatty acid synthesis is critical for the proper S-acylation of the spike protein.
Collapse
Affiliation(s)
- Katrina Mekhail
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Minhyoung Lee
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Michael Sugiyama
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Audrey Astori
- Princess Margaret Cancer Centre, University Health Network, Ontario, Canada
| | | | - Elyse Latreille
- Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Negar Khosraviani
- Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Kuiru Wei
- Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Zhijie Li
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - James Rini
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Costin Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
43
|
Lai S, Chen Y, Yang F, Xiao W, Liu Y, Wang C. Quantitative Site-Specific Chemoproteomic Profiling of Protein Lipoylation. J Am Chem Soc 2022; 144:10320-10329. [PMID: 35648456 DOI: 10.1021/jacs.2c01528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein lipoylation is an evolutionarily conserved post-translational modification from prokaryotes to eukaryotes. Lipoylation is implicated with several human diseases, including metabolic disorders, cancer, and Alzheimer's disease. While individual lipoylated proteins have been biochemically studied, a strategy for globally quantifying lipoylation with site-specific resolution in proteomes is still lacking. Herein, we developed a butyraldehyde-alkynyl probe to specifically label and enrich lipoylations in complexed biological samples. Combined with a chemoproteomic pipeline using customized tandem enzyme digestions and a biotin enrichment tag with enhanced ionization, we successfully quantified all known lipoylation sites in both Escherichia coli (E. coli) and human proteomes. The strategy enabled us to dissect the dependence of three evolutionarily related lipoylation sites in dihydrolipoamide acetyltransferase (ODP2) in E. coli and evaluated the functional connection between the de novo lipoylation synthetic pathway and the salvage pathway. Our chemoproteomic platform provides a useful tool to monitor the state of lipoylation in proteome samples, which will help decipher molecular mechanisms of lipoylation-related diseases.
Collapse
Affiliation(s)
- Shuchang Lai
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ying Chen
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Fan Yang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Weidi Xiao
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuan Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chu Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
44
|
Wen S, Song Y, Li C, Jin N, Zhai J, Lu H. Positive Regulation of the Antiviral Activity of Interferon-Induced Transmembrane Protein 3 by S-Palmitoylation. Front Immunol 2022; 13:919477. [PMID: 35769480 PMCID: PMC9236556 DOI: 10.3389/fimmu.2022.919477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
The interferon-induced transmembrane protein 3 (IFITM3), a small molecule transmembrane protein induced by interferon, is generally conserved in vertebrates, which can inhibit infection by a diverse range of pathogenic viruses such as influenza virus. However, the precise antiviral mechanisms of IFITM3 remain unclear. At least four post-translational modifications (PTMs) were found to modulate the antiviral effect of IFITM3. These include positive regulation provided by S-palmitoylation of cysteine and negative regulation provided by lysine ubiquitination, lysine methylation, and tyrosine phosphorylation. IFITM3 S-palmitoylation is an enzymatic addition of a 16-carbon fatty acid on the three cysteine residues within or adjacent to its two hydrophobic domains at positions 71, 72, and 105, that is essential for its proper targeting, stability, and function. As S-palmitoylation is the only PTM known to enhance the antiviral activity of IFITM3, enzymes that add this modification may play important roles in IFN-induced immune responses. This study mainly reviews the research progresses on the antiviral mechanism of IFITM3, the regulation mechanism of S-palmitoylation modification on its subcellular localization, stability, and function, and the enzymes that mediate the S-palmitoylation modification of IFITM3, which may help elucidate the mechanism by which this IFN effector restrict virus replication and thus aid in the design of therapeutics targeted at pathogenic viruses.
Collapse
Affiliation(s)
- Shubo Wen
- Preventive Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control, Universities of Inner Mongolia Autonomous Region, Tongliao, China
| | - Yang Song
- Preventive Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control, Universities of Inner Mongolia Autonomous Region, Tongliao, China
| | - Chang Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jingbo Zhai
- Preventive Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- Key Laboratory of Zoonose Prevention and Control, Universities of Inner Mongolia Autonomous Region, Tongliao, China
| | - Huijun Lu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
45
|
Kumar M, Carr P, Turner SR. An atlas of Arabidopsis protein S-acylation reveals its widespread role in plant cell organization and function. NATURE PLANTS 2022. [PMID: 35681017 DOI: 10.1101/2020.05.12.090415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
S-acylation is the addition of a fatty acid to a cysteine residue of a protein. While this modification may profoundly alter protein behaviour, its effects on the function of plant proteins remains poorly characterized, largely as a result of the lack of basic information regarding which proteins are S-acylated and where in the proteins the modification occurs. To address this gap in our knowledge, we used an optimized acyl-resin-assisted capture assay to perform a comprehensive analysis of plant protein S-acylation from six separate tissues. In our high- and medium-confidence groups, we identified 1,849 cysteines modified by S-acylation, which were located in 1,640 unique peptides from 1,094 different proteins. This represents around 6% of the detectable Arabidopsis proteome and suggests an important role for S-acylation in many essential cellular functions including trafficking, signalling and metabolism. To illustrate the potential of this dataset, we focus on cellulose synthesis and confirm the S-acylation of a number of proteins known to be involved in cellulose synthesis and trafficking of the cellulose synthase complex. In the secondary cell walls, cellulose synthesis requires three different catalytic subunits (CESA4, CESA7 and CESA8) that all exhibit striking sequence similarity and are all predicted to possess a RING-type zinc finger at their amino terminus composed of eight cysteines. For CESA8, we find evidence for S-acylation of these cysteines that is incompatible with any role in coordinating metal ions. We show that while CESA7 may possess a RING-type domain, the same region of CESA8 appears to have evolved a very different structure. Together, the data suggest that this study represents an atlas of S-acylation in Arabidopsis that will facilitate the broader study of this elusive post-translational modification in plants as well as demonstrating the importance of further work in this area.
Collapse
Affiliation(s)
- Manoj Kumar
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Paul Carr
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Holiferm, Manchester, UK
| | - Simon R Turner
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
46
|
Tian Y, Zeng H, Wu J, Huang J, Gao Q, Tang D, Cai L, Liao Z, Wang Y, Liu X, Lin J. Screening DHHCs of S-acylated proteins using an OsDHHC cDNA library and bimolecular fluorescence complementation in rice. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2022; 110:1763-1780. [PMID: 35411551 DOI: 10.1111/tpj.15769] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/30/2022] [Accepted: 04/07/2022] [Indexed: 05/28/2023]
Abstract
S-acylation is an important lipid modification that primarily involves DHHC proteins (DHHCs) and associated S-acylated proteins. No DHHC-S-acylated protein pair has been reported so far in rice (Oryza sativa L.) and the molecular mechanisms underlying S-acylation in plants are largely unknown. We constructed an OsDHHC cDNA library for screening corresponding pairs of DHHCs and S-acylated proteins using bimolecular fluorescence complementation assays. Five DHHC-S-acylated protein pairs (OsDHHC30-OsCBL2, OsDHHC30-OsCBL3, OsDHHC18-OsNOA1, OsDHHC13-OsNAC9, and OsDHHC14-GSD1) were identified in rice. Among the pairs, OsCBL2 and OsCBL3 were S-acylated by OsDHHC30 in yeast and rice. The localization of OsCBL2 and OsCBL3 in the endomembrane depended on S-acylation mediated by OsDHHC30. Meanwhile, all four OsDHHCs screened complemented the thermosensitive phenotype of an akr1 yeast mutant, and their DHHC motifs were required for S-acyltransferase activity. Overexpression of OsDHHC30 in rice plants improved their salt and oxidative tolerance. Together, these results contribute to our understanding of the molecular mechanism underlying S-acylation in plants.
Collapse
Affiliation(s)
- Ye Tian
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Hui Zeng
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Jicai Wu
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Jian Huang
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Qiang Gao
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Dongying Tang
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Lipeng Cai
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Zhaoyi Liao
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Yan Wang
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Xuanming Liu
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Jianzhong Lin
- Hunan Province Key Laboratory of Plant Functional Genomics and Developmental Regulation, State Key Laboratory of Chemo/Biosensing and Chemometrics, National Center of Technology Innovation for Saline-Alkali Tolerant Rice, College of Biology, Hunan University, Changsha, 410082, Hunan, China
| |
Collapse
|
47
|
Kumar M, Carr P, Turner SR. An atlas of Arabidopsis protein S-acylation reveals its widespread role in plant cell organization and function. NATURE PLANTS 2022; 8:670-681. [PMID: 35681017 PMCID: PMC7617270 DOI: 10.1038/s41477-022-01164-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/04/2022] [Indexed: 05/28/2023]
Abstract
S-acylation is the addition of a fatty acid to a cysteine residue of a protein. While this modification may profoundly alter protein behaviour, its effects on the function of plant proteins remains poorly characterized, largely as a result of the lack of basic information regarding which proteins are S-acylated and where in the proteins the modification occurs. To address this gap in our knowledge, we used an optimized acyl-resin-assisted capture assay to perform a comprehensive analysis of plant protein S-acylation from six separate tissues. In our high- and medium-confidence groups, we identified 1,849 cysteines modified by S-acylation, which were located in 1,640 unique peptides from 1,094 different proteins. This represents around 6% of the detectable Arabidopsis proteome and suggests an important role for S-acylation in many essential cellular functions including trafficking, signalling and metabolism. To illustrate the potential of this dataset, we focus on cellulose synthesis and confirm the S-acylation of a number of proteins known to be involved in cellulose synthesis and trafficking of the cellulose synthase complex. In the secondary cell walls, cellulose synthesis requires three different catalytic subunits (CESA4, CESA7 and CESA8) that all exhibit striking sequence similarity and are all predicted to possess a RING-type zinc finger at their amino terminus composed of eight cysteines. For CESA8, we find evidence for S-acylation of these cysteines that is incompatible with any role in coordinating metal ions. We show that while CESA7 may possess a RING-type domain, the same region of CESA8 appears to have evolved a very different structure. Together, the data suggest that this study represents an atlas of S-acylation in Arabidopsis that will facilitate the broader study of this elusive post-translational modification in plants as well as demonstrating the importance of further work in this area.
Collapse
Affiliation(s)
- Manoj Kumar
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Paul Carr
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Holiferm, Manchester, UK
| | - Simon R Turner
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
48
|
Wang H, Wang L, Luo X, Guan J, Zhang X, Zhang L, Xiang Y. Molecular cloning, expression and anti-tumor function analysis of yak IFITM2 gene. Int J Biol Macromol 2022; 209:405-412. [PMID: 35381283 DOI: 10.1016/j.ijbiomac.2022.03.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 11/25/2022]
Abstract
IFITM2 is interferon-induced transmembrane protein 2, which plays an extremely key role in anti-tumor and anti-virus diseases. In this study, the 602 bp cDNA sequence of the yak (Bos grunniens) IFITM2 (BgIFITM2) gene was obtained. Moreover, the prokaryotic expression vector of BgIFITM2 protein was constructed and expressed successfully, with a molecular weight of 33.680 kDa. The proliferation activities and migration abilities of HepG2 cells were significantly inhibited after treatment with BgIFITM2 protein (0.1 and 1 μg/mL) (P < 0.05). The expressions of B cell lymphoma-2 (BCL2)/BCL2-associated X (BAX) and molecular target of rapamycin (mTOR) genes were significantly decreased, but the expressions of BAX gene were significantly increased after treatment with BgIFITM2 protein (0.1 and 1 μg/mL) (P < 0.05). The expression of BAX protein was also significantly increased after treatment with 1 μg/mL BgIFITM2 protein (P < 0.05). Finally, the addition of BgIFITM2 protein attenuated the formation of tumor lesions in mice, and the pathological damage of the lung was less than that in the model group. The expression of Ki67 protein in the model group was significantly higher than that in the control group (P < 0.05), but the expression of Ki67 protein in the BgIFITM2 group was significantly lower than that in the model group (P < 0.05). Taken together, BgIFITM2 protein could inhibit the proliferative activity of HepG2 cells by regulating apoptosis-related genes, and reduce the invasiveness of HepG2 cells in mice lung tissue. These results facilitate further studies on the function of BgIFITM2 protein.
Collapse
Affiliation(s)
- Haipeng Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, PR China
| | - Li Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, PR China.
| | - Xiaolin Luo
- Sichuan Academy of Grassland Sciences, Chengdu 611731, PR China
| | - Jiuqiang Guan
- Sichuan Academy of Grassland Sciences, Chengdu 611731, PR China
| | - Xiangfei Zhang
- Sichuan Academy of Grassland Sciences, Chengdu 611731, PR China
| | - Ling Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, PR China
| | - Yi Xiang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, PR China
| |
Collapse
|
49
|
Zhang B, Li S, Shui W. Post-Translational Modifications of G Protein–Coupled Receptors Revealed by Proteomics and Structural Biology. Front Chem 2022; 10:843502. [PMID: 35355784 PMCID: PMC8960047 DOI: 10.3389/fchem.2022.843502] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/16/2022] [Indexed: 01/20/2023] Open
Abstract
G protein–coupled receptors (GPCRs) are a protein superfamily comprising >800 members that regulate numerous cellular and physiologic responses. GPCRs represent the largest class of therapeutic targets with implications in various diseases. Although advances in GPCR structural and pharmacological research have significantly improved our knowledge of GPCR signaling mechanisms, mapping diverse post-translational modifications (PTMs) of GPCR proteins and understanding their regulatory roles have received much less attention. Mass spectrometry-based proteomics has become the most popular technology for profiling protein PTMs in a systematic manner. Herein we provide an overview of PTM types, locations, crosstalk and dynamic regulation for different GPCRs that are characterized using proteomic and/or biochemical approaches. Our main focus is on glycosylation, phosphorylation, ubiquitination and palmitoylation that are known to modulate receptor folding, biosynthesis, trafficking, dimerization and signaling. Furthermore, we discuss the locations of specific PTM sites in the structure of a given GPCR and its signaling complex to highlight the importance of PTM regulation in the molecular basis of GPCRs, which may shed new light on structure-based drug discovery.
Collapse
Affiliation(s)
- Bingjie Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Shanshan Li
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- *Correspondence: Wenqing Shui,
| |
Collapse
|
50
|
Rudnik S, Heybrock S, Saftig P, Damme M. S-palmitoylation determines TMEM55B-dependent positioning of lysosomes. J Cell Sci 2022; 135:jcs258566. [PMID: 34350967 DOI: 10.1242/jcs.258566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/23/2021] [Indexed: 11/20/2022] Open
Abstract
The spatiotemporal cellular distribution of lysosomes depends on active transport mainly driven by microtubule motors such as kinesins and dynein. Different protein complexes attach these molecular motors to their vesicular cargo. TMEM55B (also known as PIP4P1), as an integral lysosomal membrane protein, is a component of such a complex that mediates the retrograde transport of lysosomes by establishing interactions with the cytosolic scaffold protein JIP4 (also known as SPAG9) and dynein-dynactin. Here, we show that TMEM55B and its paralog TMEM55A (PIP4P2) are S-palmitoylated proteins that are lipidated at multiple cysteine residues. Mutation of all cysteines in TMEM55B prevents S-palmitoylation and causes retention of the mutated protein in the Golgi. Consequently, non-palmitoylated TMEM55B is no longer able to modulate lysosomal positioning and the perinuclear clustering of lysosomes. Additional mutagenesis of the dileucine-based lysosomal sorting motif in non-palmitoylated TMEM55B leads to partial missorting to the plasma membrane instead of retention in the Golgi, implicating a direct effect of S-palmitoylation on the adaptor protein-dependent sorting of TMEM55B. Our data suggest a critical role for S-palmitoylation in the trafficking of TMEM55B and TMEM55B-dependent lysosomal positioning.
Collapse
Affiliation(s)
- Sönke Rudnik
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Saskia Heybrock
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Markus Damme
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| |
Collapse
|