1
|
Gao Y, Wei G, Yu H, Li S, Tang Y, Yue X, Chen Y, Zhan M, Wu J. Integrin β6/Annexin A2 axis triggers autophagy to orchestrate hepatocellular carcinoma radioresistance. Cell Death Differ 2025; 32:689-701. [PMID: 39533071 PMCID: PMC11982560 DOI: 10.1038/s41418-024-01411-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/26/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Radiotherapy (RT) is one of the main therapies for hepatocellular carcinoma (HCC), but its effectiveness has been constrained due to the resistance effect of radiation. Thus, the factors involved in radioresistance are evaluated and the underlying molecular mechanisms are also done. In this present study, we identified Integrin β6 (ITGB6) as a potential radioresistant gene through an integrative analysis of transcriptomic profiles, proteome datasets and survival using HCC cases treated with IR. We show that ITGB6 functionally contributed to radioresistance by activating autophagy through a series of in vitro and in vivo methods, such as clonogenic assays, autophagy flux (LC3B-GFP-mCherry reporter) analysis and a subcutaneous transplantation model. Mechanically, ITGB6 binds to Annexin A2 (ANXA2) and enhanced its stability by competitively antagonizing proteasome mediated ANXA2 degradation, thereby promoting autophagy and radioresistance. Notably, HCC radioresistance was significantly improved by either blocking ITGB6 or autophagy, but the combination was more effective. Importantly, ITGB6/ANXA2 axis triggered autophagic program endowed HCC cells with radioresistant activity in a radiated patient-derived xenograft (PDX) model and hydrodynamic injection in liver-specific Itgb6-knockout mice, further supported by clinical evidence. Together, our data revealed that ITGB6 is a radioresistant gene stabilizing the autophagy regulatory protein ANXA2, providing insights into the biological and potentially clinical significance of ITGB6/ANXA2 axis in radiotherapy planning of HCC.
Collapse
Affiliation(s)
- Ying Gao
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Cancer Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guangyan Wei
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Cancer Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hua Yu
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, China
| | - Shuping Li
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Cancer Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhao Tang
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Cancer Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xin Yue
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Cancer Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yong Chen
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Cancer Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meixiao Zhan
- Department of Interventional Medicine, Guangzhou First Pepople's Hospital, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong, China.
| | - Jian Wu
- Center of Hepato-Pancreato-Biliary Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Virard F, Giraud S, Bonnet M, Magadoux L, Martin L, Pham TH, Skafi N, Deneuve S, Frem R, Villoutreix BO, Sleiman NH, Reboulet J, Merabet S, Chaptal V, Chaveroux C, Hussein N, Aznar N, Fenouil T, Treilleux I, Saintigny P, Ansieau S, Manié S, Lebecque S, Renno T, Coste I. Targeting ERK-MYD88 interaction leads to ERK dysregulation and immunogenic cancer cell death. Nat Commun 2024; 15:7037. [PMID: 39147750 PMCID: PMC11327251 DOI: 10.1038/s41467-024-51275-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: 01/10/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024] Open
Abstract
The quest for targeted therapies is critical in the battle against cancer. The RAS/MAP kinase pathway is frequently implicated in neoplasia, with ERK playing a crucial role as the most distal kinase in the RAS signaling cascade. Our previous research demonstrated that the interaction between ERK and MYD88, an adaptor protein in innate immunity, is crucial for RAS-dependent transformation and cancer cell survival. In this study, we examine the biological consequences of disrupting the ERK-MYD88 interaction through the ERK D-recruitment site (DRS), while preserving ERK's kinase activity. Our results indicate that EI-52, a small-molecule benzimidazole targeting ERK-MYD88 interaction induces an HRI-mediated integrated stress response (ISR), resulting in immunogenic apoptosis specific to cancer cells. Additionally, EI-52 exhibits anti-tumor efficacy in patient-derived tumors and induces an anti-tumor T cell response in mice in vivo. These findings suggest that inhibiting the ERK-MYD88 interaction may be a promising therapeutic approach in cancer treatment.
Collapse
Affiliation(s)
- François Virard
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
- University of Lyon, Faculté d'Odontologie, Hospices Civils de Lyon, Lyon, France
| | - Stéphane Giraud
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
- Center for Drug Discovery and Development, Synergy Lyon Cancer Foundation, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Mélanie Bonnet
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Léa Magadoux
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Laetitia Martin
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
- Center for Drug Discovery and Development, Synergy Lyon Cancer Foundation, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Thuy Ha Pham
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Najwa Skafi
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Sophie Deneuve
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Rita Frem
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Bruno O Villoutreix
- Université de Paris, NeuroDiderot, Inserm, Hôpital Robert Debré, 75019, Paris, France
| | - Nawal Hajj Sleiman
- Institut de Génomique Fonctionnelle de Lyon, UMR 5242-CNRS/ENSL, Université Claude Bernard Lyon 1, Lyon, France
| | - Jonathan Reboulet
- Institut de Génomique Fonctionnelle de Lyon, UMR 5242-CNRS/ENSL, Université Claude Bernard Lyon 1, Lyon, France
| | - Samir Merabet
- Institut de Génomique Fonctionnelle de Lyon, UMR 5242-CNRS/ENSL, Université Claude Bernard Lyon 1, Lyon, France
| | - Vincent Chaptal
- Drug Resistance & Membrane Proteins group, Molecular Microbiology and Structural Biochemistry Laboratory (CNRS UMR 5086), University of Lyon, Lyon, France
| | - Cédric Chaveroux
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Nader Hussein
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Nicolas Aznar
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Tanguy Fenouil
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
- University of Lyon, Faculté de Médecine, Hospices Civils de Lyon, Lyon, France
| | | | - Pierre Saintigny
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Stéphane Ansieau
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Serge Manié
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Serge Lebecque
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France
- University of Lyon, Faculté de Médecine, Hospices Civils de Lyon, Lyon, France
| | - Toufic Renno
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France.
| | - Isabelle Coste
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Lyon Cancer Research Center, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
3
|
Wang W, Ouyang J, Li Y, Zhai C, He B, Si H, Chen K, Rose JKC, Jia W. A signaling cascade mediating fruit trait development via phosphorylation-modulated nuclear accumulation of JAZ repressor. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2024; 66:1106-1125. [PMID: 38558522 DOI: 10.1111/jipb.13654] [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: 12/12/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
It is generally accepted that jasmonate-ZIM domain (JAZ) repressors act to mediate jasmonate (JA) signaling via CORONATINE-INSENSITIVE1 (COI1)-mediated degradation. Here, we report a cryptic signaling cascade where a JAZ repressor, FvJAZ12, mediates multiple signaling inputs via phosphorylation-modulated subcellular translocation rather than the COI1-mediated degradation mechanism in strawberry (Fragaria vesca). FvJAZ12 acts to regulate flavor metabolism and defense response, and was found to be the target of FvMPK6, a mitogen-activated protein kinase that is capable of responding to multiple signal stimuli. FvMPK6 phosphorylates FvJAZ12 at the amino acid residues S179 and T183 adjacent to the PY residues, thereby attenuating its nuclear accumulation and relieving its repression for FvMYC2, which acts to control the expression of lipoxygenase 3 (FvLOX3), an important gene involved in JA biosynthesis and a diverse array of cellular metabolisms. Our data reveal a previously unreported mechanism for JA signaling and decipher a signaling cascade that links multiple signaling inputs with fruit trait development.
Collapse
Affiliation(s)
- Wei Wang
- College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Jinyao Ouyang
- College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Yating Li
- College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Changsheng Zhai
- College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Bing He
- College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Huahan Si
- College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Kunsong Chen
- College of Agriculture and Biotechnology, Zhejiang University, Zijingang Campus, Hangzhou, 310058, China
| | - Jocelyn K C Rose
- Plant Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, 14853, NY, USA
| | - Wensuo Jia
- College of Horticulture, China Agricultural University, Beijing, 100193, China
- Institute of Horticulture Crops, Xinjiang Academy of Agricultural Sciences, Urumqi, 830000, China
| |
Collapse
|
4
|
Chen X, Leyendecker S. Kinematic analysis of kinases and their oncogenic mutations - Kinases and their mutation kinematic analysis. Mol Inform 2024; 43:e202300250. [PMID: 38850084 DOI: 10.1002/minf.202300250] [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: 09/19/2023] [Revised: 01/25/2024] [Accepted: 03/14/2024] [Indexed: 06/09/2024]
Abstract
Protein kinases are crucial cellular enzymes that facilitate the transfer of phosphates from adenosine triphosphate (ATP) to their substrates, thereby regulating numerous cellular activities. Dysfunctional kinase activity often leads to oncogenic conditions. Chosen by using structural similarity to 5UG9, we selected 79 crystal structures from the PDB and based on the position of the phenylalanine side chain in the DFG motif, we classified these 79 crystal structures into 5 group clusters. Our approach applies our kinematic flexibility analysis (KFA) to explore the flexibility of kinases in various activity states and examine the impact of the activation loop on kinase structure. KFA enables the rapid decomposition of macromolecules into different flexibility regions, allowing comprehensive analysis of conformational structures. The results reveal that the activation loop of kinases acts as a "lock" that stabilizes the active conformation of kinases by rigidifying the adjacent α-helices. Furthermore, we investigate specific kinase mutations, such as the L858R mutation commonly associated with non-small cell lung cancer, which induces increased flexibility in active-state kinases. In addition, through analyzing the hydrogen bond pattern, we examine the substructure of kinases in different states. Notably, active-state kinases exhibit a higher occurrence of α-helices compared to inactive-state kinases. This study contributes to the understanding of biomolecular conformation at a level relevant to drug development.
Collapse
Affiliation(s)
- Xiyu Chen
- Institute of Applied Dynamics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Sigrid Leyendecker
- Institute of Applied Dynamics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
| |
Collapse
|
5
|
Zhang Z, Gao X, Tian Z, Yang E, Huang Y, Liu D, Dai S, Zhang H, Bao M, Jiang X, Li X, Luo P. Preso enhances mGluR1-mediated excitotoxicity by modulating the phosphorylation of mGluR1-Homer1 complex and facilitating an ER stress after traumatic brain injury. Cell Death Discov 2024; 10:153. [PMID: 38531909 DOI: 10.1038/s41420-024-01916-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/10/2023] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Glutamate receptor (GluR)-mediated excitotoxicity is an important mechanism causing delayed neuronal injury after traumatic brain injury (TBI). Preso, as a core scaffolding protein of postsynaptic density (PSD), is considered an important regulator during excitotoxicity and TBI and combines with glutamate receptors to form functional units for excitatory glutamatergic neurotransmission, and elucidating the mechanisms of these functional units will provide new targets for the treatment of TBI. As a multidomain scaffolding protein, Preso directly interacts with metabotropic GluR (mGluR) and another scaffold protein, Homer. Because the mGluR-Homer complex plays a crucial role in TBI, modulation of this complex by Preso may be an important mechanism affecting the excitotoxic damage to neurons after TBI. Here, we demonstrate that Preso facilitates the interaction between metabotropic mGluR1 and Homer1 to activate mGluR1 signaling and cause excitotoxic neuronal injury and endoplasmic reticulum (ER) stress after TBI. The regulatory effect of Preso on the mGluR1-Homer1 complex is dependent on the direct association between Preso and this complex and also involves the phosphorylation of the interactive binding sites of mGluR1 and Homer1 by Preso. Further studies confirmed that Preso, as an adaptor of cyclin-dependent kinase 5 (CDK5), promotes the phosphorylation of the Homer1-binding site on mGluR1 by CDK5 and thereby enhances the interaction between mGluR1 and Homer1. Preso can also promote the formation of the mGluR1-Homer1 complex by inhibiting the phosphorylation of the Homer1 hinge region by Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα). Based on these molecular mechanisms, we designed several blocking peptides targeting the interaction between Preso and the mGluR1-Homer1 complex and found that directly disrupting the association between mGluR1 and scaffolding proteins significantly promotes the recovery of motor function after TBI.
Collapse
Affiliation(s)
- Zhuoyuan Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- School of Life Science, Northwest University, Xi'an, China
| | - Xiangyu Gao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhicheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erwan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yutao Huang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dan Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- School of Life Science, Northwest University, Xi'an, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Haofuzi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mingdong Bao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Xin Li
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
6
|
Iglesias-Romero AB, Soto T, Flor-Parra I, Salas-Pino S, Ruiz-Romero G, Gould KL, Cansado J, Daga RR. MAPK-dependent control of mitotic progression in S. pombe. BMC Biol 2024; 22:71. [PMID: 38523261 PMCID: PMC10962199 DOI: 10.1186/s12915-024-01865-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/08/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND Mitogen-activated protein kinases (MAPKs) preserve cell homeostasis by transducing physicochemical fluctuations of the environment into multiple adaptive responses. These responses involve transcriptional rewiring and the regulation of cell cycle transitions, among others. However, how stress conditions impinge mitotic progression is largely unknown. The mitotic checkpoint is a surveillance mechanism that inhibits mitotic exit in situations of defective chromosome capture, thus preventing the generation of aneuploidies. In this study, we investigate the role of MAPK Pmk1 in the regulation of mitotic exit upon stress. RESULTS We show that Schizosaccharomyces pombe cells lacking Pmk1, the MAP kinase effector of the cell integrity pathway (CIP), are hypersensitive to microtubule damage and defective in maintaining a metaphase arrest. Epistasis analysis suggests that Pmk1 is involved in maintaining spindle assembly checkpoint (SAC) signaling, and its deletion is additive to the lack of core SAC components such as Mad2 and Mad3. Strikingly, pmk1Δ cells show up to twofold increased levels of the anaphase-promoting complex (APC/C) activator Cdc20Slp1 during unperturbed growth. We demonstrate that Pmk1 physically interacts with Cdc20Slp1 N-terminus through a canonical MAPK docking site. Most important, the Cdc20Slp1 pool is rapidly degraded in stressed cells undergoing mitosis through a mechanism that requires MAPK activity, Mad3, and the proteasome, thus resulting in a delayed mitotic exit. CONCLUSIONS Our data reveal a novel function of MAPK in preventing mitotic exit and activation of cytokinesis in response to stress. The regulation of Cdc20Slp1 turnover by MAPK Pmk1 provides a key mechanism by which the timing of mitotic exit can be adjusted relative to environmental conditions.
Collapse
Affiliation(s)
| | - Terersa Soto
- Yeast Physiology Group, Department of Genetics and Microbiology, Facultad de Biología, Universidad de Murcia, Murcia, 30071, Spain
| | - Ignacio Flor-Parra
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain
| | - Silvia Salas-Pino
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain
| | - Gabriel Ruiz-Romero
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - José Cansado
- Yeast Physiology Group, Department of Genetics and Microbiology, Facultad de Biología, Universidad de Murcia, Murcia, 30071, Spain.
| | - Rafael R Daga
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Seville, 41013, Spain.
| |
Collapse
|
7
|
Zerihun M, Rubin SJS, Silnitsky S, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part II: Peptides as Allosteric Protein Kinase C Modulators Targeting Protein-Protein Interactions. Int J Mol Sci 2023; 24:17504. [PMID: 38139336 PMCID: PMC10743673 DOI: 10.3390/ijms242417504] [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: 11/01/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Human protein kinases are highly-sought-after drug targets, historically harnessed for treating cancer, cardiovascular disease, and an increasing number of autoimmune and inflammatory conditions. Most current treatments involve small molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP-binding pocket. As a result, these compounds are often poorly selective and highly toxic. Part I of this series reviews the role of PKC isoforms in various human diseases, featuring cancer and cardiovascular disease, as well as translational examples of PKC modulation applied to human health and disease. In the present Part II, we discuss alternative allosteric binding mechanisms for targeting PKC, as well as novel drug platforms, such as modified peptides. A major goal is to design protein kinase modulators with enhanced selectivity and improved pharmacological properties. To this end, we use molecular docking analysis to predict the mechanisms of action for inhibitor-kinase interactions that can facilitate the development of next-generation PKC modulators.
Collapse
Affiliation(s)
- Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed 1311502, Israel; (M.Z.); (S.S.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed 1311502, Israel; (M.Z.); (S.S.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed 1311502, Israel; (M.Z.); (S.S.)
| |
Collapse
|
8
|
Wilcockson SG, Guglielmi L, Araguas Rodriguez P, Amoyel M, Hill CS. An improved Erk biosensor detects oscillatory Erk dynamics driven by mitotic erasure during early development. Dev Cell 2023; 58:2802-2818.e5. [PMID: 37714159 PMCID: PMC7615346 DOI: 10.1016/j.devcel.2023.08.021] [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: 11/01/2022] [Revised: 06/02/2023] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
Extracellular signal-regulated kinase (Erk) signaling dynamics elicit distinct cellular responses in a variety of contexts. The early zebrafish embryo is an ideal model to explore the role of Erk signaling dynamics in vivo, as a gradient of activated diphosphorylated Erk (P-Erk) is induced by fibroblast growth factor (Fgf) signaling at the blastula margin. Here, we describe an improved Erk-specific biosensor, which we term modified Erk kinase translocation reporter (modErk-KTR). We demonstrate the utility of this biosensor in vitro and in developing zebrafish and Drosophila embryos. Moreover, we show that Fgf/Erk signaling is dynamic and coupled to tissue growth during both early zebrafish and Drosophila development. Erk activity is rapidly extinguished just prior to mitosis, which we refer to as mitotic erasure, inducing periods of inactivity, thus providing a source of heterogeneity in an asynchronously dividing tissue. Our modified reporter and transgenic lines represent an important resource for interrogating the role of Erk signaling dynamics in vivo.
Collapse
Affiliation(s)
- Scott G Wilcockson
- Developmental Signalling Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Luca Guglielmi
- Developmental Signalling Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Pablo Araguas Rodriguez
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Marc Amoyel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
9
|
Jin H, Zhang W, Liu H, Bao Y. Genome-wide identification and characteristic analysis of ETS gene family in blood clam Tegillarca granosa. BMC Genomics 2023; 24:700. [PMID: 37990147 PMCID: PMC10664356 DOI: 10.1186/s12864-023-09731-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/11/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND ETS transcription factors, known as the E26 transformation-specific factors, assume a critical role in the regulation of various vital biological processes in animals, including cell differentiation, the cell cycle, and cell apoptosis. However, their characterization in mollusks is currently lacking. RESULTS The current study focused on a comprehensive analysis of the ETS genes in blood clam Tegillarca granosa and other mollusk genomes. Our phylogenetic analysis revealed the absence of the SPI and ETV subfamilies in mollusks compared to humans. Additionally, several ETS genes in mollusks were found to lack the PNT domain, potentially resulting in a diminished ability of ETS proteins to bind target genes. Interestingly, the bivalve ETS1 genes exhibited significantly high expression levels during the multicellular proliferation stage and in gill tissues. Furthermore, qRT-PCR results showed that Tg-ETS-14 (ETS1) is upregulated in the high total hemocyte counts (THC) population of T. granosa, suggesting it plays a significant role in stimulating hemocyte proliferation. CONCLUSION Our study significantly contributes to the comprehension of the evolutionary aspects concerning the ETS gene family, while also providing valuable insights into its role in fostering hemocyte proliferation across mollusks.
Collapse
Affiliation(s)
- Hongyu Jin
- School of Marine Sciences, Ningbo University, Ningbo, 315000, China
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Zhejiang, 315100, China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, 315000, China
| | - Hongxing Liu
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Zhejiang, 315100, China.
| | - Yongbo Bao
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Zhejiang, 315100, China.
| |
Collapse
|
10
|
Chen X, Li Q, Ding L, Zhang S, Shan S, Xiong X, Jiang W, Zhao B, Zhang L, Luo Y, Lian Y, Kong X, Ding X, Zhang J, Li C, Soppe WJJ, Xiang Y. The MKK3-MPK7 cascade phosphorylates ERF4 and promotes its rapid degradation to release seed dormancy in Arabidopsis. MOLECULAR PLANT 2023; 16:1743-1758. [PMID: 37710960 DOI: 10.1016/j.molp.2023.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/18/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
Seeds establish dormancy to delay germination until the arrival of a favorable growing season. In this study, we identify a fate switch comprised of the MKK3-MPK7 kinase cascade and the ethylene response factor ERF4 that is responsible for the seed state transition from dormancy to germination. We show that dormancy-breaking factors activate the MKK3-MPK7 module, which affects the expression of some α-EXPANSIN (EXPA) genes to control seed dormancy. Furthermore, we identify a direct downstream substrate of this module, ERF4, which suppresses the expression of these EXPAs by directly binding to the GCC boxes in their exon regions. The activated MKK3-MPK7 module phosphorylates ERF4, leading to its rapid degradation and thereby releasing its inhibitory effect on the expression of these EXPAs. Collectively, our work identifies a signaling chain consisting of protein phosphorylation, degradation, and gene transcription , by which the germination promoters within the embryo sense and are activated by germination signals from ambient conditions.
Collapse
Affiliation(s)
- Xi Chen
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Qiujia Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Ling Ding
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Shengnan Zhang
- Center for Crop Science, College of Agronomy, Qingdao Agricultural University, Qingdao 266109, China
| | - Siyao Shan
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Xiong Xiong
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Wenhui Jiang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Bo Zhao
- Hou Ji Laboratory in Shanxi Province, Academy of Agronomy, Shanxi Agricultural University, Taiyuan 030031, China
| | - Liying Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Ying Luo
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Yiming Lian
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Xiuqin Kong
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Xiali Ding
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Jun Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | - Chunli Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China
| | | | - Yong Xiang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China.
| |
Collapse
|
11
|
Lin CY, Zhang YM, Li BZ, Shu MA, Xu WB. Identification and characterization of mitogen-activated protein kinase kinase 4 (MKK4) from the mud crab Scylla paramamosain in response to Vibrio alginolyticus and White Spot Syndrome Virus (WSSV). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104755. [PMID: 37295629 DOI: 10.1016/j.dci.2023.104755] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
Mitogen-activated protein kinase kinase 4 (MKK4), serves as a critical component of the mitogen-activated protein kinase signaling pathway, facilitating the direct phosphorylation and activation of the c-Jun N-terminal kinase (JNK) and p38 families of MAP kinases in response to environmental stresses. In the current research, we identified two MKK4 subtypes, namely SpMKK4-1 and SpMKK4-2, from Scylla paramamosain, followed by the analysis of their molecular characteristics and tissue distributions. The expression of SpMKK4s was induced upon WSSV and Vibrio alginolyticus challenges, and the bacteria clearance capacity and antimicrobial peptide (AMP) genes' expression upon bacterial infection were significantly decreased after knocking down SpMKK4s. Additionally, the overexpression of both SpMKK4s remarkably activated NF-κB reporter plasmid in HEK293T cells, suggesting the activation of the NF-κB signaling pathway. These results indicated the participation of SpMKK4s in the innate immunity of crabs, which shed light on a better understanding of the mechanisms through which MKK4s regulate innate immunity.
Collapse
Affiliation(s)
- Chen-Yang Lin
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yan-Mei Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bang-Ze Li
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Miao-An Shu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Wen-Bin Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
12
|
Yan Y, Jiang JY, Fu M, Wang D, Pelletier AR, Sigdel D, Ng DC, Wang W, Ping P. MIND-S is a deep-learning prediction model for elucidating protein post-translational modifications in human diseases. CELL REPORTS METHODS 2023; 3:100430. [PMID: 37056379 PMCID: PMC10088250 DOI: 10.1016/j.crmeth.2023.100430] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/19/2023] [Accepted: 02/24/2023] [Indexed: 03/29/2023]
Abstract
We present a deep-learning-based platform, MIND-S, for protein post-translational modification (PTM) predictions. MIND-S employs a multi-head attention and graph neural network and assembles a 15-fold ensemble model in a multi-label strategy to enable simultaneous prediction of multiple PTMs with high performance and computation efficiency. MIND-S also features an interpretation module, which provides the relevance of each amino acid for making the predictions and is validated with known motifs. The interpretation module also captures PTM patterns without any supervision. Furthermore, MIND-S enables examination of mutation effects on PTMs. We document a workflow, its applications to 26 types of PTMs of two datasets consisting of ∼50,000 proteins, and an example of MIND-S identifying a PTM-interrupting SNP with validation from biological data. We also include use case analyses of targeted proteins. Taken together, we have demonstrated that MIND-S is accurate, interpretable, and efficient to elucidate PTM-relevant biological processes in health and diseases.
Collapse
Affiliation(s)
- Yu Yan
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Medical Informatics Program, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Physiology, UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr., Los Angeles, CA 90095-1760, USA
| | - Jyun-Yu Jiang
- Scalable Analytics Institute (ScAi) at Department of Computer Science, UCLA School of Engineering, Los Angeles, CA 90095, USA
| | - Mingzhou Fu
- Medical Informatics Program, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Ding Wang
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Department of Physiology, UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr., Los Angeles, CA 90095-1760, USA
| | - Alexander R. Pelletier
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Department of Physiology, UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr., Los Angeles, CA 90095-1760, USA
- Scalable Analytics Institute (ScAi) at Department of Computer Science, UCLA School of Engineering, Los Angeles, CA 90095, USA
| | - Dibakar Sigdel
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Department of Physiology, UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr., Los Angeles, CA 90095-1760, USA
| | - Dominic C.M. Ng
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Department of Physiology, UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr., Los Angeles, CA 90095-1760, USA
| | - Wei Wang
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Medical Informatics Program, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Scalable Analytics Institute (ScAi) at Department of Computer Science, UCLA School of Engineering, Los Angeles, CA 90095, USA
| | - Peipei Ping
- NIH BRIDGE2AI Center at UCLA & NHLBI Integrated Cardiovascular Data Science Training Program at UCLA, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
- Medical Informatics Program, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Physiology, UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr., Los Angeles, CA 90095-1760, USA
- Scalable Analytics Institute (ScAi) at Department of Computer Science, UCLA School of Engineering, Los Angeles, CA 90095, USA
- Department of Medicine (Cardiology), UCLA School of Medicine, Suite 1-609, MRL Building, 675 Charles E. Young Dr. South, Los Angeles, CA 90095-1760, USA
| |
Collapse
|
13
|
Dan Y, Radic N, Gay M, Fernández-Torras A, Arauz G, Vilaseca M, Aloy P, Canovas B, Nebreda A. Characterization of p38α signaling networks in cancer cells using quantitative proteomics and phosphoproteomics. Mol Cell Proteomics 2023; 22:100527. [PMID: 36894123 PMCID: PMC10105487 DOI: 10.1016/j.mcpro.2023.100527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
p38α (encoded by MAPK14) is a protein kinase that regulates cellular responses to almost all types of environmental and intracellular stresses. Upon activation, p38α phosphorylates many substrates both in the cytoplasm and nucleus, allowing this pathway to regulate a wide variety of cellular processes. While the role of p38α in the stress response has been widely investigated, its implication in cell homeostasis is less understood. To investigate the signaling networks regulated by p38α in proliferating cancer cells, we performed quantitative proteomic and phosphoproteomic analyses in breast cancer cells in which this pathway had been either genetically targeted or chemically inhibited. Our study identified with high confidence 35 proteins and 82 phosphoproteins (114 phosphosites) that are modulated by p38α, and highlighted the implication of various protein kinases, including MK2 and mTOR, in the p38α-regulated signaling networks. Moreover, functional analyses revealed an important contribution of p38α to the regulation of cell adhesion, DNA replication and RNA metabolism. Indeed, we provide experimental evidence supporting that p38α facilitates cancer cell adhesion, and showed that this p38α function is likely mediated by the modulation of the adaptor protein ArgBP2. Collectively, our results illustrate the complexity of the p38α regulated signaling networks, provide valuable information on p38α-dependent phosphorylation events in cancer cells, and document a mechanism by which p38α can regulate cell adhesion.
Collapse
Affiliation(s)
- Yuzhen Dan
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Nevenka Radic
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Marina Gay
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Adrià Fernández-Torras
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Gianluca Arauz
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Marta Vilaseca
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - AngelR Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
14
|
Shi G, Song C, Torres Robles J, Salichos L, Lou HJ, Lam TT, Gerstein M, Turk BE. Proteome-wide screening for mitogen-activated protein kinase docking motifs and interactors. Sci Signal 2023; 16:eabm5518. [PMID: 36626580 PMCID: PMC9995140 DOI: 10.1126/scisignal.abm5518] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Essential functions of mitogen-activated protein kinases (MAPKs) depend on their capacity to selectively phosphorylate a limited repertoire of substrates. MAPKs harbor a conserved groove located outside of the catalytic cleft that binds to short linear sequence motifs found in substrates and regulators. However, the weak and transient nature of these "docking" interactions poses a challenge to defining MAPK interactomes and associated sequence motifs. Here, we describe a yeast-based genetic screening pipeline to evaluate large collections of MAPK docking sequences in parallel. Using this platform, we analyzed a combinatorial library based on the docking sequences from the MAPK kinases MKK6 and MKK7, defining features critical for binding to the stress-activated MAPKs JNK1 and p38α. Our screen of a library consisting of ~12,000 sequences from the human proteome revealed multiple MAPK-selective interactors, including many that did not conform to previously defined docking motifs. Analysis of p38α/JNK1 exchange mutants identified specific docking groove residues that mediate selective binding. Last, we verified that docking sequences identified in the screen functioned in substrate recruitment in vitro and in cultured cells. Together, these studies establish an approach to characterize MAPK docking sequences and provide a resource for future investigation of signaling downstream of p38 and JNK.
Collapse
Affiliation(s)
- Guangda Shi
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Claire Song
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jaylissa Torres Robles
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Leonidas Salichos
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Hua Jane Lou
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA.,Keck MS and Proteomics Resource, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mark Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Benjamin E Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
15
|
Zhang J, Pei J, Durham J, Bos T, Cong Q. Computed cancer interactome explains the effects of somatic mutations in cancers. Protein Sci 2022; 31:e4479. [PMID: 36261849 PMCID: PMC9667826 DOI: 10.1002/pro.4479] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 12/13/2022]
Abstract
Protein-protein interactions (PPIs) are involved in almost all essential cellular processes. Perturbation of PPI networks plays critical roles in tumorigenesis, cancer progression, and metastasis. While numerous high-throughput experiments have produced a vast amount of data for PPIs, these data sets suffer from high false positive rates and exhibit a high degree of discrepancy. Coevolution of amino acid positions between protein pairs has proven to be useful in identifying interacting proteins and providing structural details of the interaction interfaces with the help of deep learning methods like AlphaFold (AF). In this study, we applied AF to investigate the cancer protein-protein interactome. We predicted 1,798 PPIs for cancer driver proteins involved in diverse cellular processes such as transcription regulation, signal transduction, DNA repair, and cell cycle. We modeled the spatial structures for the predicted binary protein complexes, 1,087 of which lacked previous 3D structure information. Our predictions offer novel structural insight into many cancer-related processes such as the MAP kinase cascade and Fanconi anemia pathway. We further investigated the cancer mutation landscape by mapping somatic missense mutations (SMMs) in cancer to the predicted PPI interfaces and performing enrichment and depletion analyses. Interfaces enriched or depleted with SMMs exhibit different preferences for functional categories. Interfaces enriched in mutations tend to function in pathways that are deregulated in cancers and they may help explain the molecular mechanisms of cancers in patients; interfaces lacking mutations appear to be essential for the survival of cancer cells and thus may be future targets for PPI modulating drugs.
Collapse
Affiliation(s)
- Jing Zhang
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jimin Pei
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jesse Durham
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Tasia Bos
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
16
|
Barnhart MD, Yang Y, Nakagaki-Silva EE, Hammond TH, Pizzinga M, Gooding C, Stott K, Smith CWJ. Phosphorylation of the smooth muscle master splicing regulator RBPMS regulates its splicing activity. Nucleic Acids Res 2022; 50:11895-11915. [PMID: 36408906 PMCID: PMC9723635 DOI: 10.1093/nar/gkac1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/12/2022] [Accepted: 10/24/2022] [Indexed: 11/22/2022] Open
Abstract
We previously identified RBPMS as a master regulator of alternative splicing in differentiated smooth muscle cells (SMCs). RBPMS is transcriptionally downregulated during SMC dedifferentiation, but we hypothesized that RBPMS protein activity might be acutely downregulated by post-translational modifications. Publicly available phosphoproteomic datasets reveal that Thr113 and Thr118 immediately adjacent to the RRM domain are commonly both phosphorylated. An RBPMS T113/118 phosphomimetic T/E mutant showed decreased splicing regulatory activity both in transfected cells and in a cell-free in vitro assay, while a non-phosphorylatable T/A mutant retained full activity. Loss of splicing activity was associated with a modest reduction in RNA affinity but significantly reduced RNA binding in nuclear extract. A lower degree of oligomerization of the T/E mutant might cause lower avidity of multivalent RNA binding. However, NMR analysis also revealed that the T113/118E peptide acts as an RNA mimic which can loop back and antagonize RNA-binding by the RRM domain. Finally, we identified ERK2 as the most likely kinase responsible for phosphorylation at Thr113 and Thr118. Collectively, our data identify a potential mechanism for rapid modulation of the SMC splicing program in response to external signals during the vascular injury response and atherogenesis.
Collapse
Affiliation(s)
- Michael D Barnhart
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Yi Yang
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | | - Thomas H Hammond
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | | - Clare Gooding
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Katherine Stott
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | |
Collapse
|
17
|
Solomon O, Shpilt Z, Sapir H, Marom S, Bibas S, Chen Y, Tshuva EY, Yitzchaik S, Friedler A. Peptide‐Based Inhibitors that Target the Docking Site of ERK2. Isr J Chem 2022. [DOI: 10.1002/ijch.202200041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Ohad Solomon
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
- Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Zohar Shpilt
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Hannah Sapir
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
- Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Shir Marom
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
- Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Shai Bibas
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
- Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Yu‐Ju Chen
- Institute of Chemistry Academia Sinica No. 128, Section2, Academia Road Taipei 115 Taiwan
| | - Edit Y. Tshuva
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Shlomo Yitzchaik
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
- Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| | - Assaf Friedler
- Institute of Chemistry, T he Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
- Center for Nanoscience and Nanotechnology The Hebrew University of Jerusalem Safra Campus, Givat Ram Jerusalem 91904 Israel
| |
Collapse
|
18
|
Athanasopoulou K, Adamopoulos PG, Scorilas A. Structural characterization and expression analysis of novel MAPK1 transcript variants with the development of a multiplexed targeted nanopore sequencing approach. Int J Biochem Cell Biol 2022; 150:106272. [PMID: 35878809 DOI: 10.1016/j.biocel.2022.106272] [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: 02/08/2022] [Revised: 06/17/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) represent a protein family firmly involved in many signaling cascades, regulating a vast spectrum of stimulated cellular processes. Studies have shown that alternatively spliced isoforms of MAPKs play a crucial role in determining the desired cell fate in response to specific stimulations. Although the implication of most MAPKs transcript variants in the MAPK signaling cascades has been clarified, the transcriptional profile of a pivotal member, MAPK1, has not been investigated for the existence of additional isoforms. In the current study we developed and implemented targeted long-read and short-read sequencing approaches to identify novel MAPK1 splice variants. The combination of nanopore sequencing and NGS enabled the implementation of a long-read polishing pipeline using error-rate correction algorithms, which empowered the high accuracy of the results and increased the sequencing efficiency. The utilized multiplexing option in the nanopore sequencing approach allowed not only the identification of novel MAPK1 mRNAs, but also elucidated their expression profile in multiple human malignancies and non-cancerous cell lines. Our study highlights for the first time the existence of ten previously undescribed MAPK1 mRNAs (MAPK1 v.3 - v.12) and evaluates their relative expression levels in comparison to the main MAPK1 v.1. The optimization and employment of qPCR assays revealed that MAPK1 v.3 - v.12 can be quantified in a wide spectrum of human cell lines with notable specificity. Finally, our findings suggest that the novel protein-coding mRNAs are highly expected to participate in the regulation of MAPK pathways, demonstrating differential localizations and functionalities.
Collapse
Affiliation(s)
- Konstantina Athanasopoulou
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
19
|
Solone XKV, Caldara AL, Wells B, Qiao H, Wade LR, Salerno JC, Helms KA, Smith KER, McMurry JL, Chrestensen CA. MAP kinases differentially bind and phosphorylate NOS3 via two unique NOS3 sites. FEBS Open Bio 2022; 12:1075-1086. [PMID: 35182051 PMCID: PMC9063426 DOI: 10.1002/2211-5463.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/12/2022] [Accepted: 02/17/2022] [Indexed: 11/25/2022] Open
Abstract
Nitric oxide synthase 3 (NOS3) is a major vasoprotective enzyme that catalyzes the conversion of l-arginine to nitric oxide (NO) in response to a significant number of signaling pathways. Here, we provide evidence that NOS3 interactions with MAP kinases have physiological relevance. Binding interactions of NOS3 with c-Jun N-terminal kinase (JNK1α1 ), p38α, and ERK2 were characterized using optical biosensing with full-length NOS3 and NOS3 specific peptides and phosphopeptides. Like p38α and ERK2, JNK1α1 exhibited high-affinity binding to full-length NOS3 (KD 15 nm). Rate constants exhibited fast-on, slow-off binding (kon = 4106 m-1 s-1 ; koff = 6.2 × 10-5 s-1 ). Further analysis using synthetic NOS3 peptides revealed two MAP kinase binding sites unique to NOS3. p38α evinced similar affinity with both NOS3 binding sites. For ERK2 and JNK1α1, the affinity at the two sites differed. However, NOS3 peptides with a phosphate at either S114 or S633 did not meaningfully interact with the kinases. Immunoblotting revealed that each kinase phosphorylated NOS3 with a unique pattern. JNK1α1 predominantly phosphorylated NOS3 at S114, ERK2 at S600, and p38α phosphorylated both residues. In vitro production of NO was unchanged by phosphorylation at these sites. In human microvascular endothelial cells, endogenous interactions of all the MAP kinases with NOS3 were captured using proximity ligation assay in resting cells. Our results underscore the importance of MAP kinase interactions, identifying two unique NOS3 interaction sites with potential for modulation by MAP kinase phosphorylation (S114) and other signaling inputs, like protein kinase A (S633).
Collapse
Affiliation(s)
- Xzaviar K. V. Solone
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
- Present address:
Department of Molecular Genetics and MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Amber L. Caldara
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
| | - Brady Wells
- Department of Chemistry & BiochemistryKennesaw State UniversityGAUSA
| | - Hao Qiao
- Department of Chemistry & BiochemistryKennesaw State UniversityGAUSA
| | - Lydia R. Wade
- Department of Chemistry & BiochemistryKennesaw State UniversityGAUSA
| | - John C. Salerno
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
| | - Katy A. Helms
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
- Present address:
Wake Forest Medical CenterWinston‐SalemNCUSA
| | | | | | | |
Collapse
|
20
|
Sun T, Zhang Y. MAP kinase cascades in plant development and immune signaling. EMBO Rep 2022; 23:e53817. [PMID: 35041234 PMCID: PMC8811656 DOI: 10.15252/embr.202153817] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/26/2021] [Accepted: 01/01/2022] [Indexed: 02/05/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) cascades are important signaling modules regulating diverse biological processes. During the past 20 years, much progress has been made on the functions of MAPK cascades in plants. This review summarizes the roles of MAPKs, known MAPK substrates, and our current understanding of MAPK cascades in plant development and innate immunity. In addition, recent findings on the molecular links connecting surface receptors to MAPK cascades and the mechanisms underlying MAPK signaling specificity are also discussed.
Collapse
Affiliation(s)
- Tongjun Sun
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Yuelin Zhang
- Department of BotanyUniversity of British ColumbiaVancouverBCCanada
| |
Collapse
|
21
|
Lee CJ, Lee GE, An HJ, Cho ES, Chen W, Lee JY, Kang HC, Lee HS, Cho YY. F-box Protein βTrCP1 Is a Substrate of Extracellular Signal-regulated Kinase 2. J Cancer Prev 2021; 26:174-182. [PMID: 34703820 PMCID: PMC8511579 DOI: 10.15430/jcp.2021.26.3.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 11/28/2022] Open
Abstract
F-box proteins, consisting of 69 members which are organized into the three subclasses FBXW, FBXL, and FBXO, are the substrate specific recognition subunits of the SKP1-Cullin 1-F-box protein E3 ligase complex. Although βTrCP 1 and 2, members of the FBXW subfamily, are known to regulate some protein stability, molecular mechanisms by which these proteins can recognize proper substrates are unknown. In this study, it was found that βTrCP1 showed strong interaction with members of mitogen-activated protein kinases. Although extracellular signal-regulated kinase (ERK) 3, p38β, and p38δ showed weak interactions, ERK2 specifically interacted with βTrCP1 as assessed by immunoprecipitation. In interaction domain determination experiments, we found that ERK2 interacted with two independent ERK docking sites located in the F-box domain and linker domain, but not the WD40 domain, of βTrCP1. Notably, mutations of βTrCP1 at the ERK docking sites abolished the interaction with ERK2. βTrCP1 underwent phosphorylation by EGF stimulation, while the presence of the mitogen-activated protein kinase kinases inhibitor U0126, genetic silencing by sh-ERK2, and mutation of the ERK docking site of βTrCP1 inhibited phosphorylation. This inhibition of βTrCP1 phosphorylation resulted in a shortened half-life and low protein levels. These results suggest that ERK2-mediated βTrCP1 phosphorylation may induce the destabilization of βTrCP1.
Collapse
Affiliation(s)
- Cheol-Jung Lee
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea.,Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon, Korea
| | - Ga-Eun Lee
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Hyun-Jung An
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Eun Suh Cho
- Department of Biochemistry, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Weidong Chen
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Joo Young Lee
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Han Chang Kang
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Hye Suk Lee
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Yong-Yeon Cho
- BRL & BK21-4th Team, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| |
Collapse
|
22
|
Yan Z, Wang J, Wang F, Xie C, Lv B, Yu Z, Dai S, Liu X, Xia G, Tian H, Li C, Ding Z. MPK3/6-induced degradation of ARR1/10/12 promotes salt tolerance in Arabidopsis. EMBO Rep 2021; 22:e52457. [PMID: 34402578 DOI: 10.15252/embr.202152457] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/18/2021] [Accepted: 07/30/2021] [Indexed: 11/09/2022] Open
Abstract
Cytokinins are phytohormones that regulate plant development, growth, and responses to stress. In particular, cytokinin has been reported to negatively regulate plant adaptation to high salinity; however, the molecular mechanisms that counteract cytokinin signaling and enable salt tolerance are not fully understood. Here, we provide evidence that salt stress induces the degradation of the cytokinin signaling components Arabidopsis (Arabidopisis thaliana) response regulator 1 (ARR1), ARR10 and ARR12. Furthermore, the stress-activated mitogen-activated protein kinase 3 (MPK3) and MPK6 interact with and phosphorylate ARR1/10/12 to promote their degradation in response to salt stress. As expected, salt tolerance is decreased in the mpk3/6 double mutant, but enhanced upon ectopic MPK3/MPK6 activation in an MKK5DD line. Importantly, salt hypersensitivity phenotypes of the mpk3/6 line were significantly alleviated by mutation of ARR1/12. The above results indicate that MPK3/6 enhance salt tolerance in part via their negative regulation of ARR1/10/12 protein stability. Thus, our work reveals a new molecular mechanism underlying salt-induced stress adaptation and the inhibition of plant growth, via enhanced degradation of cytokinin signaling components.
Collapse
Affiliation(s)
- Zhenwei Yan
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Junxia Wang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Fengxia Wang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Chuantian Xie
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Bingsheng Lv
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Zipeng Yu
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Shaojun Dai
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Xia Liu
- Maize Research Institute, Shandong Academy of Agricultural Sciences/National Engineering Laboratory of Wheat and Maize/Key Laboratory of Biology and Genetic Improvement of Maize in Northern Yellow-huai River Plain, Ministry of Agriculture, Jinan, China
| | - Guangmin Xia
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Huiyu Tian
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Cuiling Li
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhaojun Ding
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, China
| |
Collapse
|
23
|
Paes D, Schepers M, Rombaut B, van den Hove D, Vanmierlo T, Prickaerts J. The Molecular Biology of Phosphodiesterase 4 Enzymes as Pharmacological Targets: An Interplay of Isoforms, Conformational States, and Inhibitors. Pharmacol Rev 2021; 73:1016-1049. [PMID: 34233947 DOI: 10.1124/pharmrev.120.000273] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The phosphodiesterase 4 (PDE4) enzyme family plays a pivotal role in regulating levels of the second messenger cAMP. Consequently, PDE4 inhibitors have been investigated as a therapeutic strategy to enhance cAMP signaling in a broad range of diseases, including several types of cancers, as well as in various neurologic, dermatological, and inflammatory diseases. Despite their widespread therapeutic potential, the progression of PDE4 inhibitors into the clinic has been hampered because of their related relatively small therapeutic window, which increases the chance of producing adverse side effects. Interestingly, the PDE4 enzyme family consists of several subtypes and isoforms that can be modified post-translationally or can engage in specific protein-protein interactions to yield a variety of conformational states. Inhibition of specific PDE4 subtypes, isoforms, or conformational states may lead to more precise effects and hence improve the safety profile of PDE4 inhibition. In this review, we provide an overview of the variety of PDE4 isoforms and how their activity and inhibition is influenced by post-translational modifications and interactions with partner proteins. Furthermore, we describe the importance of screening potential PDE4 inhibitors in view of different PDE4 subtypes, isoforms, and conformational states rather than testing compounds directed toward a specific PDE4 catalytic domain. Lastly, potential mechanisms underlying PDE4-mediated adverse effects are outlined. In this review, we illustrate that PDE4 inhibitors retain their therapeutic potential in myriad diseases, but target identification should be more precise to establish selective inhibition of disease-affected PDE4 isoforms while avoiding isoforms involved in adverse effects. SIGNIFICANCE STATEMENT: Although the PDE4 enzyme family is a therapeutic target in an extensive range of disorders, clinical use of PDE4 inhibitors has been hindered because of the adverse side effects. This review elaborately shows that safer and more effective PDE4 targeting is possible by characterizing 1) which PDE4 subtypes and isoforms exist, 2) how PDE4 isoforms can adopt specific conformations upon post-translational modifications and protein-protein interactions, and 3) which PDE4 inhibitors can selectively bind specific PDE4 subtypes, isoforms, and/or conformations.
Collapse
Affiliation(s)
- Dean Paes
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Melissa Schepers
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Ben Rombaut
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Daniel van den Hove
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Tim Vanmierlo
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Jos Prickaerts
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| |
Collapse
|
24
|
Im JH, Ko JH, Kim WC, Crain B, Keathley D, Han KH. Mitogen-activated protein kinase 6 negatively regulates secondary wall biosynthesis by modulating MYB46 protein stability in Arabidopsis thaliana. PLoS Genet 2021; 17:e1009510. [PMID: 33826618 PMCID: PMC8055014 DOI: 10.1371/journal.pgen.1009510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 04/19/2021] [Accepted: 03/25/2021] [Indexed: 11/18/2022] Open
Abstract
The R2R3-MYB transcription factor MYB46 functions as a master switch for secondary cell wall biosynthesis, ensuring the exquisite expression of the secondary wall biosynthetic genes in the tissues where secondary walls are critical for growth and development. At the same time, suppression of its function is needed when/where formation of secondary walls is not desirable. Little is known about how this opposing control of secondary cell wall formation is achieved. We used both transient and transgenic expression of MYB46 and mitogen-activated protein kinase 6 (MPK6) to investigate the molecular mechanism of the post-translational regulation of MYB46. We show that MYB46 is phosphorylated by MPK6, leading to site specific phosphorylation-dependent degradation of MYB46 by the ubiquitin-mediated proteasome pathway. In addition, the MPK6-mediated MYB46 phosphorylation was found to regulate in planta secondary wall forming function of MYB46. Furthermore, we provide experimental evidences that MYB83, a paralog of MYB46, is not regulated by MPK6. The coupling of MPK signaling to MYB46 function provides insights into the tissue- and/or condition-specific activity of MYB46 for secondary wall biosynthesis.
Collapse
Affiliation(s)
- Jong Hee Im
- Department of Horticulture, Michigan State University, East Lansing, Michigan, United States of America
- National Academy of Agricultural Science, Rural Development Administration, Jeonju, Republic of Korea
| | - Jae-Heung Ko
- Department of Plant & Environmental New Resources, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea
- * E-mail: (J-HK); (K-HH)
| | - Won-Chan Kim
- School of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Brent Crain
- Department of Horticulture, Michigan State University, East Lansing, Michigan, United States of America
| | - Daniel Keathley
- Department of Horticulture, Michigan State University, East Lansing, Michigan, United States of America
| | - Kyung-Hwan Han
- Department of Horticulture, Michigan State University, East Lansing, Michigan, United States of America
- Department of Forestry, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail: (J-HK); (K-HH)
| |
Collapse
|
25
|
Lv B, Wei K, Hu K, Tian T, Zhang F, Yu Z, Zhang D, Su Y, Sang Y, Zhang X, Ding Z. MPK14-mediated auxin signaling controls lateral root development via ERF13-regulated very-long-chain fatty acid biosynthesis. MOLECULAR PLANT 2021; 14:285-297. [PMID: 33221411 DOI: 10.1016/j.molp.2020.11.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 11/15/2020] [Indexed: 05/16/2023]
Abstract
Auxin plays a critical role in lateral root (LR) formation. The signaling module composed of auxin-response factors (ARFs) and lateral organ boundaries domain transcription factors mediates auxin signaling to control almost every stage of LR development. Here, we show that auxin-induced degradation of the APETALA2/Ethylene Responsive Factor (AP2/ERF) transcription factor ERF13, dependent on MITOGEN-ACTIVATED PROTEIN KINASE MPK14-mediated phosphorylation, plays an essential role in LR development. Overexpression of ERF13 results in restricted passage of the LR primordia through the endodermal layer, greatly reducing LR emergence, whereas the erf13 mutants showed an increase in emerged LR. ERF13 inhibits the expression of 3-ketoacyl-CoA synthase16 (KCS16), which encodes a fatty acid elongase involved in very-long-chain fatty acid (VLCFA) biosynthesis. Overexpression of KCS16 or exogenous VLCFA treatment rescues the LR emergence defects in ERF13 overexpression lines, indicating a role downstream of the auxin-MPK14-ERF13 signaling module. Collectively, our study uncovers a novel molecular mechanism by which MPK14-mediated auxin signaling modulates LR development via ERF13-regulated VLCFA biosynthesis.
Collapse
Affiliation(s)
- Bingsheng Lv
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China; College of Horticulture, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Kaijing Wei
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Kongqin Hu
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Te Tian
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Feng Zhang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Zipeng Yu
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Dajian Zhang
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yinghua Su
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yalin Sang
- State Key Laboratory of Crop Biology, College of Forestry, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Xiansheng Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhaojun Ding
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
26
|
Park HC, Park BO, Kim HS, Kim SH, Lee SW, Chung WS. AtMPK6-induced phosphorylation of AtERF72 enhances its DNA binding activity and interaction with TGA4/OBF4 in Arabidopsis. PLANT BIOLOGY (STUTTGART, GERMANY) 2021; 23:11-20. [PMID: 33073469 DOI: 10.1111/plb.13196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/09/2020] [Indexed: 06/11/2023]
Abstract
The ethylene-responsive element binding factor (ERF) family is a large family of transcription factors involved in plant development and environmental stress responses. We previously reported the identification of 29 putative substrates of Mitogen-activated Protein Kinase3 (AtMPK3), AtMPK4 and AtMPK6, based on a solid-phase phosphorylation screening using a lambda phage expression library in Arabidopsis thaliana. In this study, a putative MPK substrate, AtERF72 (At3g16770), was strongly phosphorylated by AtMPK6 on the serine residue at position 151 (Ser151). AtERF72 binds to the GCC box (AGCCGCC) in the promoters of several pathogenesis-related (PR) genes and activates their transcription. We also show that the DNA-binding activity of AtERF72 is enhanced upon phosphorylation by AtMPK6 in vitro. In addition, transient co-expression experiments in Arabidopsis protoplasts revealed that effector constructs expressing a mutant variant of AtERF72, AtERF72S151D (carrying a Ser to aspartic acid [Asp] substitution at amino acid position 151) showed higher expression of the β-glucuronidase (GUS) reporter gene driven by the GCC box element than effector constructs expressing the wild-type AtERF72. Furthermore, yeast two-hybrid assays revealed that the interaction between AtERF72S151D and TGA4/OBF4 was stronger than that between wild-type AtERF72 and TGA4/OBF4. Since AtERF72S151D is equivalent to AtERF72 phosphorylated by AtMPK6 at Ser151, these results suggest that the phosphorylation of AtERF72 by AtMPK6 triggers an event of transcriptional regulation from defence signalling in Arabidopsis.
Collapse
Affiliation(s)
- H C Park
- Team of Vulnerable Ecological Research, Division of Climate and Ecology, Bureau of Conservation & Assessment Research, National Institute of Ecology, Seocheon, Republic of Korea
| | - B O Park
- Division of Applied Life Science (BK21 Plus Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, Republic of Korea
| | - H S Kim
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - S H Kim
- Division of Applied Life Science (BK21 Plus Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, Republic of Korea
| | - S W Lee
- Department of Agronomy & Medicinal Plant Resources, Gyeongnam National University of Science & Technology, Jinju, Republic of Korea
| | - W S Chung
- Division of Applied Life Science (BK21 Plus Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
27
|
Russo R, Mallia S, Zito F, Lampiasi N. Long-Lasting Activity of ERK Kinase Depends on NFATc1 Induction and Is Involved in Cell Migration-Fusion in Murine Macrophages RAW264.7. Int J Mol Sci 2020; 21:ijms21238965. [PMID: 33255852 PMCID: PMC7728313 DOI: 10.3390/ijms21238965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Macrophages are mononuclear cells that become osteoclasts (OCs) in the presence of two cytokines, macrophage colony-stimulating factor (M-CSF), and receptor activator of NF-κB ligand (RANKL). RANKL binding to its specific receptor RANK leads to OCs differentiation mainly by nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). In our previous study, the analysis of the protein network in NFATc1-knockdown cells, using the Ingenuity Pathway Analysis (IPA), showed a link between NFATc1 and Mitogen-activated protein kinase kinase (MEK)-extracellular receptor kinase (ERK) signaling pathway. Therefore, this study aimed to extend our knowledge of the relationship between NFATc1 and the ERK. Here, we demonstrate that delayed ERK1/2 phosphorylation in pre-OC RANKL-induced depends on NFATc1. Indeed, the knockdown of NFATc1 reduced the phosphorylation of ERK1/2 (60%) and the pharmacological inhibition of the ERK1/2 kinase activity impairs the expression of NFATc1 without preventing its translocation into the nucleus. Furthermore, silencing of NFATc1 significantly reduced RANKL-induced migration (p < 0.01), and most pre-OCs are still mononuclear after 48 h (80 ± 5%), despite the presence of actin rings. On the other hand, the inhibitors FR180204 and PD98059 significantly reduced RANKL-induced cell migration (p < 0.01), leading to a reduction in the number of multinucleated cells. Finally, we suggest that long-lasting ERK activity depends on NFATc1 induction and is likely linked to cell migration, fusion, and OC differentiation.
Collapse
|
28
|
Kirsch K, Zeke A, Tőke O, Sok P, Sethi A, Sebő A, Kumar GS, Egri P, Póti ÁL, Gooley P, Peti W, Bento I, Alexa A, Reményi A. Co-regulation of the transcription controlling ATF2 phosphoswitch by JNK and p38. Nat Commun 2020; 11:5769. [PMID: 33188182 PMCID: PMC7666158 DOI: 10.1038/s41467-020-19582-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/21/2020] [Indexed: 01/26/2023] Open
Abstract
Transcription factor phosphorylation at specific sites often activates gene expression, but how environmental cues quantitatively control transcription is not well-understood. Activating protein 1 transcription factors are phosphorylated by mitogen-activated protein kinases (MAPK) in their transactivation domains (TAD) at so-called phosphoswitches, which are a hallmark in response to growth factors, cytokines or stress. We show that the ATF2 TAD is controlled by functionally distinct signaling pathways (JNK and p38) through structurally different MAPK binding sites. Moreover, JNK mediated phosphorylation at an evolutionarily more recent site diminishes p38 binding and made the phosphoswitch differently sensitive to JNK and p38 in vertebrates. Structures of MAPK-TAD complexes and mechanistic modeling of ATF2 TAD phosphorylation in cells suggest that kinase binding motifs and phosphorylation sites line up to maximize MAPK based co-regulation. This study shows how the activity of an ancient transcription controlling phosphoswitch became dependent on the relative flux of upstream signals.
Collapse
Affiliation(s)
- Klára Kirsch
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - András Zeke
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - Orsolya Tőke
- Laboratory for NMR Spectroscopy, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - Péter Sok
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - Ashish Sethi
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anna Sebő
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | | | - Péter Egri
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - Ádám L Póti
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - Paul Gooley
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wolfgang Peti
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Isabel Bento
- European Molecular Biology Laboratory, Hamburg, Germany
| | - Anita Alexa
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary
| | - Attila Reményi
- Biomolecular Interactions Research Group, Institute of Organic Chemistry, Research Center for Natural Sciences, H-1117, Budapest, Hungary.
| |
Collapse
|
29
|
Motta M, Pannone L, Pantaleoni F, Bocchinfuso G, Radio FC, Cecchetti S, Ciolfi A, Di Rocco M, Elting MW, Brilstra EH, Boni S, Mazzanti L, Tamburrino F, Walsh L, Payne K, Fernández-Jaén A, Ganapathi M, Chung WK, Grange DK, Dave-Wala A, Reshmi SC, Bartholomew DW, Mouhlas D, Carpentieri G, Bruselles A, Pizzi S, Bellacchio E, Piceci-Sparascio F, Lißewski C, Brinkmann J, Waclaw RR, Waisfisz Q, van Gassen K, Wentzensen IM, Morrow MM, Álvarez S, Martínez-García M, De Luca A, Memo L, Zampino G, Rossi C, Seri M, Gelb BD, Zenker M, Dallapiccola B, Stella L, Prada CE, Martinelli S, Flex E, Tartaglia M. Enhanced MAPK1 Function Causes a Neurodevelopmental Disorder within the RASopathy Clinical Spectrum. Am J Hum Genet 2020; 107:499-513. [PMID: 32721402 DOI: 10.1016/j.ajhg.2020.06.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/24/2020] [Indexed: 12/23/2022] Open
Abstract
Signal transduction through the RAF-MEK-ERK pathway, the first described mitogen-associated protein kinase (MAPK) cascade, mediates multiple cellular processes and participates in early and late developmental programs. Aberrant signaling through this cascade contributes to oncogenesis and underlies the RASopathies, a family of cancer-prone disorders. Here, we report that de novo missense variants in MAPK1, encoding the mitogen-activated protein kinase 1 (i.e., extracellular signal-regulated protein kinase 2, ERK2), cause a neurodevelopmental disease within the RASopathy phenotypic spectrum, reminiscent of Noonan syndrome in some subjects. Pathogenic variants promote increased phosphorylation of the kinase, which enhances translocation to the nucleus and boosts MAPK signaling in vitro and in vivo. Two variant classes are identified, one of which directly disrupts binding to MKP3, a dual-specificity protein phosphatase negatively regulating ERK function. Importantly, signal dysregulation driven by pathogenic MAPK1 variants is stimulus reliant and retains dependence on MEK activity. Our data support a model in which the identified pathogenic variants operate with counteracting effects on MAPK1 function by differentially impacting the ability of the kinase to interact with regulators and substrates, which likely explains the minor role of these variants as driver events contributing to oncogenesis. After nearly 20 years from the discovery of the first gene implicated in Noonan syndrome, PTPN11, the last tier of the MAPK cascade joins the group of genes mutated in RASopathies.
Collapse
|
30
|
Ranasinghe ADCU, Lee DD, Schwarz MA. Mechanistic regulation of SPHK1 expression and translocation by EMAP II in pulmonary smooth muscle cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158789. [PMID: 32771459 DOI: 10.1016/j.bbalip.2020.158789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/22/2020] [Accepted: 08/02/2020] [Indexed: 12/20/2022]
Abstract
Phosphorylation of sphingosine by sphingosine kinase 1 (SPHK1) produces the bioactive sphingolipid sphingosine-1-phosphate (S1P), a microvascular and immuno-modulator associated with vascular remodeling in pulmonary arterial hypertension (PAH). The low intracellular concentration of S1P is under tight spatial-temporal control. Molecular mechanisms that mediate S1P burden and S1P regulation of vascular remodeling are poorly understood. Similarities between two early response pro-inflammatory cytokine gene transcript activation profiles, S1P and Endothelial Monocyte Activating Polypeptide II (EMAP II), suggested a strategic link between their signaling pathways. We determined that EMAP II triggers a bimodal phosphorylation, transcriptional regulation and membrane translocation of SPHK1 through a common upstream process in both macrophages and pulmonary artery smooth muscle cells (PASMCs). EMAP II initiates a dual function of ERK1/2: phosphorylation of SPHK1 and regulation of the transcription factor EGR1 that induces expression of SPHK1. Activated ERK1/2 induces a bimodal phosphorylation of SPHK1 which reciprocally increases S1P levels. This identified common upstream signaling mechanism between a protein and a bioactive lipid initiates cell specific downstream signaling representing a multifactorial mechanism that contributes to inflammation and PASMC proliferation which are cardinal histopathological phenotypes of PAH.
Collapse
Affiliation(s)
- A Dushani C U Ranasinghe
- Harper Cancer Research Institute, USA; Department of Chemistry and Biochemistry, University of Notre Dame, USA
| | - Daniel D Lee
- Harper Cancer Research Institute, USA; Departments of Pediatrics and Anatomy, Cell Biology & Physiology, Indiana University, South Bend, IN, USA
| | - Margaret A Schwarz
- Harper Cancer Research Institute, USA; Department of Chemistry and Biochemistry, University of Notre Dame, USA; Departments of Pediatrics and Anatomy, Cell Biology & Physiology, Indiana University, South Bend, IN, USA.
| |
Collapse
|
31
|
Paul S, Yang L, Mattingly H, Goyal Y, Shvartsman SY, Veraksa A. Activation-induced substrate engagement in ERK signaling. Mol Biol Cell 2020; 31:235-243. [PMID: 31913744 PMCID: PMC7183763 DOI: 10.1091/mbc.e19-07-0355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The extracellular signal-regulated kinase (ERK) pathway is an essential component of developmental signaling in metazoans. Previous models of pathway activation suggested that dissociation of activated dually phosphorylated ERK (dpERK) from MAPK/ERK kinase (MEK), a kinase that phosphorylates ERK, and other cytoplasmic anchors, is sufficient for allowing ERK interactions with its substrates. Here, we provide evidence for an additional step controlling ERK’s access to substrates. Specifically, we demonstrate that interaction of ERK with its substrate Capicua (Cic) is controlled at the level of ERK phosphorylation, whereby Cic binds to dpERK much stronger than to unphosphorylated ERK, both in vitro and in vivo. Mathematical modeling suggests that the differential affinity of Cic for dpERK versus ERK is required for both down-regulation of Cic and stabilizing phosphorylated ERK. Preferential association of Cic with dpERK serves two functions: it prevents unproductive competition of Cic with unphosphorylated ERK and contributes to efficient signal propagation. We propose that high-affinity substrate binding increases the specificity and efficiency of signal transduction through the ERK pathway.
Collapse
Affiliation(s)
- Sayantanee Paul
- Department of Biology, University of Massachusetts, Boston, Boston, MA 02125
| | - Liu Yang
- Department of Biology, University of Massachusetts, Boston, Boston, MA 02125.,Lewis-Sigler Institute for Integrative Genomics
| | - Henry Mattingly
- Lewis-Sigler Institute for Integrative Genomics.,Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544
| | - Yogesh Goyal
- Lewis-Sigler Institute for Integrative Genomics.,Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544
| | - Stanislav Y Shvartsman
- Lewis-Sigler Institute for Integrative Genomics.,Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544.,Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Alexey Veraksa
- Department of Biology, University of Massachusetts, Boston, Boston, MA 02125
| |
Collapse
|
32
|
Kaoud TS, Johnson WH, Ebelt ND, Piserchio A, Zamora-Olivares D, Van Ravenstein SX, Pridgen JR, Edupuganti R, Sammons R, Cano M, Warthaka M, Harger M, Tavares CDJ, Park J, Radwan MF, Ren P, Anslyn EV, Tsai KY, Ghose R, Dalby KN. Modulating multi-functional ERK complexes by covalent targeting of a recruitment site in vivo. Nat Commun 2019; 10:5232. [PMID: 31745079 PMCID: PMC6863825 DOI: 10.1038/s41467-019-12996-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 10/10/2019] [Indexed: 12/31/2022] Open
Abstract
Recently, the targeting of ERK with ATP-competitive inhibitors has emerged as a potential clinical strategy to overcome acquired resistance to BRAF and MEK inhibitor combination therapies. In this study, we investigate an alternative strategy of targeting the D-recruitment site (DRS) of ERK. The DRS is a conserved region that lies distal to the active site and mediates ERK-protein interactions. We demonstrate that the small molecule BI-78D3 binds to the DRS of ERK2 and forms a covalent adduct with a conserved cysteine residue (C159) within the pocket and disrupts signaling in vivo. BI-78D3 does not covalently modify p38MAPK, JNK or ERK5. BI-78D3 promotes apoptosis in BRAF inhibitor-naive and resistant melanoma cells containing a BRAF V600E mutation. These studies provide the basis for designing modulators of protein-protein interactions involving ERK, with the potential to impact ERK signaling dynamics and to induce cell cycle arrest and apoptosis in ERK-dependent cancers.
Collapse
Affiliation(s)
- Tamer S Kaoud
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA.,Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - William H Johnson
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nancy D Ebelt
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrea Piserchio
- Department of Chemistry and Biochemistry, The City College of New York, New York, NY, USA
| | | | - Sabrina X Van Ravenstein
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jacey R Pridgen
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ramakrishna Edupuganti
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Rachel Sammons
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Micael Cano
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Mangalika Warthaka
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Matthew Harger
- Biomedical Engineering Department, The University of Texas at Austin, Austin, TX, USA
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jihyun Park
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed F Radwan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Pengyu Ren
- Biomedical Engineering Department, The University of Texas at Austin, Austin, TX, USA
| | - Eric V Anslyn
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Ranajeet Ghose
- Department of Chemistry and Biochemistry, The City College of New York, New York, NY, USA.,Graduate Programs in Biochemistry, Chemistry and Physics, The Graduate Center of the City University of New York, New York, NY, 10016, USA
| | - Kevin N Dalby
- Division of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
33
|
Miao L, Tian H. Development of ERK1/2 inhibitors as a therapeutic strategy for tumour with MAPK upstream target mutations. J Drug Target 2019; 28:154-165. [PMID: 31340679 DOI: 10.1080/1061186x.2019.1648477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylate a variety of substrates that play key roles in promoting cell survival and proliferation. Many inhibitors, acting on upstream of the ERK pathway, exhibit excellent antitumor activity. However, drug-resistant tumour cells invariably emerge after their use due to the reactivation of ERK1/2 signalling. ERK1/2 inhibitors have shown clinical efficacy as a therapeutic strategy for the treatment of tumours with mitogen-activated protein kinase (MAPK) upstream target mutations. These inhibitors may be effective against cancers with altered MAPK upstream pathway and may be used as a possible strategy to overcome acquired resistance to MAPK inhibitors. In this review, we describe the mechanism and types of ERK1/2 inhibitors, summarise the current development status of small-molecule ERK1/2 inhibitors, including the preclinical data and clinical study progress, and discuss the future research directions for the application of ERK1/2 inhibitors.
Collapse
Affiliation(s)
- Longfei Miao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
34
|
Jenardhanan P, Panneerselvam M, Mathur PP. Targeting Kinase Interaction Networks: A New Paradigm in PPI Based Design of Kinase Inhibitors. Curr Top Med Chem 2019; 19:467-485. [PMID: 31184298 DOI: 10.2174/1568026619666190304155711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/20/2019] [Accepted: 02/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Kinases are key modulators in regulating diverse range of cellular activities and are an essential part of the protein-protein interactome. Understanding the interaction of kinases with different substrates and other proteins is vital to decode the cell signaling machinery as well as causative mechanism for disease onset and progression. OBJECTIVE The objective of this review is to present all studies on the structure and function of few important kinases and highlight the protein-protein interaction (PPI) mechanism of kinases and the kinase specific interactome databases and how such studies could be utilized to develop anticancer drugs. METHODS The article is a review of the detailed description of the various domains in kinases that are involved in protein-protein interactions and specific inhibitors developed targeting these PPI domains. RESULTS The review has surfaced in depth the interacting domains in key kinases and their features and the roles of PPI in the human kinome and the various signaling cascades that are involved in certain types of cancer. CONCLUSION The insight availed into the mechanism of existing peptide inhibitors and peptidomimetics against kinases will pave way for the design and generation of domain specific peptide inhibitors with better productivity and efficiency and the various software and servers available can be of great use for the identification and analysis of protein-protein interactions.
Collapse
Affiliation(s)
| | - Manivel Panneerselvam
- Department of Biotechnology, BJM School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Premendu P Mathur
- Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| |
Collapse
|
35
|
Misiura M, Kolomeisky AB. Kinetic network model to explain gain-of-function mutations in ERK2 enzyme. J Chem Phys 2019; 150:155101. [PMID: 31005085 DOI: 10.1063/1.5088647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
ERK2 is a kinase protein that belongs to a Ras/Raf/MEK/ERK signaling pathway, which is activated in response to a range of extracellular signals. Malfunctioning of this cascade leads to a variety of serious diseases, including cancers. This is often caused by mutations in proteins belonging to the cascade, frequently leading to abnormally high activity of the cascade even in the absence of an external signal. One such "gain-of-function" mutation in the ERK2 protein, called a "sevenmaker" mutation (D319N), was discovered in 1994 in Drosophila. The mutation leads to disruption of interactions of other proteins with the D-site of ERK2 and results, contrary to expectations, in an increase of its activity in vivo. However, no molecular mechanism to explain this effect has been presented so far. The difficulty is that this mutation should equally negatively affect interactions of ERK2 with all substrates, activators, and deactivators. In this paper, we present a semiquantitative kinetic network model that gives a possible explanation of the increased activity of mutant ERK2 species. A simplified biochemical network for ERK2, viewed as a system of coupled Michaelis-Menten processes, is presented. Its dynamic properties are calculated explicitly using the method of first-passage processes. The effect of mutation is associated with changes in the strength of interaction energy between the enzyme and the substrates. It is found that the dependence of kinetic properties of the protein on the interaction energy is nonmonotonic, suggesting that some mutations might lead to more efficient catalytic properties, despite weakening intermolecular interactions. Our theoretical predictions agree with experimental observations for the sevenmaker mutation in ERK2. It is also argued that the effect of mutations might depend on the concentrations of substrates.
Collapse
Affiliation(s)
- Mikita Misiura
- Department of Chemistry, Rice University, Houston, Texas 77005-1892, USA
| | | |
Collapse
|
36
|
Hiatt SM, Thompson ML, Prokop JW, Lawlor JMJ, Gray DE, Bebin EM, Rinne T, Kempers M, Pfundt R, van Bon BW, Mignot C, Nava C, Depienne C, Kalsner L, Rauch A, Joset P, Bachmann-Gagescu R, Wentzensen IM, McWalter K, Cooper GM. Deleterious Variation in BRSK2 Associates with a Neurodevelopmental Disorder. Am J Hum Genet 2019; 104:701-708. [PMID: 30879638 PMCID: PMC6451696 DOI: 10.1016/j.ajhg.2019.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/01/2019] [Indexed: 01/08/2023] Open
Abstract
Developmental delay and intellectual disability (DD and ID) are heterogeneous phenotypes that arise in many rare monogenic disorders. Because of this rarity, developing cohorts with enough individuals to robustly identify disease-associated genes is challenging. Social-media platforms that facilitate data sharing among sequencing labs can help to address this challenge. Through one such tool, GeneMatcher, we identified nine DD- and/or ID-affected probands with a rare, heterozygous variant in the gene encoding the serine/threonine-protein kinase BRSK2. All probands have a speech delay, and most present with intellectual disability, motor delay, behavioral issues, and autism. Six of the nine variants are predicted to result in loss of function, and computational modeling predicts that the remaining three missense variants are damaging to BRSK2 structure and function. All nine variants are absent from large variant databases, and BRSK2 is, in general, relatively intolerant to protein-altering variation among humans. In all six probands for whom parents were available, the mutations were found to have arisen de novo. Five of these de novo variants were from cohorts with at least 400 sequenced probands; collectively, the cohorts span 3,429 probands, and the observed rate of de novo variation in these cohorts is significantly higher than the estimated background-mutation rate (p = 2.46 × 10-6). We also find that exome sequencing provides lower coverage and appears less sensitive to rare variation in BRSK2 than does genome sequencing; this fact most likely reduces BRSK2's visibility in many clinical and research sequencing efforts. Altogether, our results implicate damaging variation in BRSK2 as a source of neurodevelopmental disease.
Collapse
Affiliation(s)
- Susan M Hiatt
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Jeremy W Prokop
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI 48824, USA
| | - James M J Lawlor
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - David E Gray
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - E Martina Bebin
- Department of Neurology, University of Alabama Birmingham, Birmingham, AL 35294, USA
| | - Tuula Rinne
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Marlies Kempers
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Bregje W van Bon
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Cyril Mignot
- Département de Génétique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris 75013, France; Centres de Référence Maladies Rares, Déficiences Intellectuelles de Causes Rares, Paris 75013, France; Groupes de Recherche Clinique Paris Sorbonne Déficience Intellectuelle et Autisme, Paris 75013, France
| | - Caroline Nava
- Département de Génétique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris 75013, France; Faculté de Médecine, Institut du Cerveau et de la Moelle épinière, Sorbonne Université, Paris 75013, France
| | - Christel Depienne
- Faculté de Médecine, Institut du Cerveau et de la Moelle épinière, Sorbonne Université, Paris 75013, France; Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Louisa Kalsner
- Connecticut Children's Medical Center, Farmington, CT 06032, USA
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren 8952, Switzerland; Radiz-Rare Disease Initiative Zurich, Clinical Research Priority Program, University of Zurich, Zurich 8032, Switzerland
| | - Pascal Joset
- Institute of Medical Genetics, University of Zurich, Schlieren 8952, Switzerland
| | | | | | | | - Gregory M Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA.
| |
Collapse
|
37
|
An In Vitro Kinase Assay to Assess Rac1 Phosphorylation by ERK. Methods Mol Biol 2019; 1821:131-140. [PMID: 30062409 DOI: 10.1007/978-1-4939-8612-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Recent findings suggest that phosphorylation might further contribute to the tight regulation of Rho GTPases. Interestingly, sequence analysis of Rac1 shows that T108 within the 106PNTP109 motif of Rac1 is likely an ERK phosphorylation site and Rac1 also has an ERK docking site 183KKRKRKCLLL192 (D-site) at the C-terminus. Protein phosphorylation could be assayed by many different methods. Here, we describe an in vitro kinase assay we used to assess Rac1 phosphorylation by ERK. Rac1 phosphorylation is detected based on the transfer of a radiolabeled phosphate from ATP to Rac1 by the phosphotransferase activity of the kinase EKR. This in vitro kinase assay uses commercially available purified active ERK. Substrate Rac1 was generated and purified as a glutathione S-transferase (GST) fusion protein. [γ-32P]ATP is used to radiolabel Rac1. Phosphorylation of Rac1 is viewed by autoradiography.
Collapse
|
38
|
Kalapos B, Hlavová M, Nádai TV, Galiba G, Bišová K, Dóczi R. Early Evolution of the Mitogen-Activated Protein Kinase Family in the Plant Kingdom. Sci Rep 2019; 9:4094. [PMID: 30858468 PMCID: PMC6411719 DOI: 10.1038/s41598-019-40751-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/04/2019] [Indexed: 01/23/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways are central cellular signalling mechanisms in all eukaryotes. They are key regulators of the cell cycle and stress responses, yet evolution of MAPK families took markedly different paths in the animal and plant kingdoms. Instead of the characteristic divergence of MAPK types in animals, in plants an expanded network of ERK-like MAPKs has emerged. To gain insight into the early evolution of the plant MAPK family we identified and analysed MAPKs in 13 representative species across green algae, a large and diverse early-diverging lineage within the plant kingdom. Our results reveal that the plant MAPK gene family emerged from three types of progenitor kinases, which are ubiquitously present in algae, implying their formation in an early ancestor. Low number of MAPKs is characteristic across algae, the few losses or duplications are associated with genome complexity rather than habitat ecology, despite the importance of MAPKs in environmental signalling in flowering plants. ERK-type MAPKs are associated with cell cycle regulation in opisthokont models, yet in plants their stress-signalling function is more prevalent. Unicellular microalgae offer an excellent experimental system to study the cell cycle, and MAPK gene expression profiles show CDKB-like peaks around S/M phase in synchronised Chlamydomonas reinhardtii cultures, suggesting their participation in cell cycle regulation, in line with the notion that the ancestral eukaryotic MAPK was a cell cycle regulator ERK-like kinase. Our work also highlights the scarcity of signalling knowledge in microalgae, in spite of their enormous ecological impact and emerging economic importance.
Collapse
Affiliation(s)
- Balázs Kalapos
- Institute of Agriculture, Centre for Agricultural Research of the Hungarian Academy of Sciences, H-2462, Martonvásár, Brunszvik u. 2, Hungary.,Festetics Doctoral School, Georgikon Faculty, University of Pannonia, 8360, Keszthely, Hungary
| | - Monika Hlavová
- Centre Algatech, Institute of Microbiology Academy of Sciences of the Czech Republic, Opatovicky mlyn, CZ 379 81, Třeboň, Czech Republic
| | - Tímea V Nádai
- Institute of Agriculture, Centre for Agricultural Research of the Hungarian Academy of Sciences, H-2462, Martonvásár, Brunszvik u. 2, Hungary.,Festetics Doctoral School, Georgikon Faculty, University of Pannonia, 8360, Keszthely, Hungary
| | - Gábor Galiba
- Institute of Agriculture, Centre for Agricultural Research of the Hungarian Academy of Sciences, H-2462, Martonvásár, Brunszvik u. 2, Hungary.,Festetics Doctoral School, Georgikon Faculty, University of Pannonia, 8360, Keszthely, Hungary
| | - Kateřina Bišová
- Centre Algatech, Institute of Microbiology Academy of Sciences of the Czech Republic, Opatovicky mlyn, CZ 379 81, Třeboň, Czech Republic
| | - Róbert Dóczi
- Institute of Agriculture, Centre for Agricultural Research of the Hungarian Academy of Sciences, H-2462, Martonvásár, Brunszvik u. 2, Hungary.
| |
Collapse
|
39
|
Clinical genome sequencing uncovers potentially targetable truncations and fusions of MAP3K8 in spitzoid and other melanomas. Nat Med 2019; 25:597-602. [PMID: 30833747 DOI: 10.1038/s41591-019-0373-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 01/22/2019] [Indexed: 12/18/2022]
Abstract
Spitzoid melanoma is a specific morphologic variant of melanoma that most commonly affects children and adolescents, and ranges on the spectrum of malignancy from low grade to overtly malignant. These tumors are generally driven by fusions of ALK, RET, NTRK1/3, MET, ROS1 and BRAF1,2. However, in approximately 50% of cases no genetic driver has been established2. Clinical whole-genome and transcriptome sequencing (RNA-Seq) of a spitzoid tumor from an adolescent revealed a novel gene fusion of MAP3K8, encoding a serine-threonine kinase that activates MEK3,4. The patient, who had exhausted all other therapeutic options, was treated with a MEK inhibitor and underwent a transient clinical response. We subsequently analyzed spitzoid tumors from 49 patients by RNA-Seq and found in-frame fusions or C-terminal truncations of MAP3K8 in 33% of cases. The fusion transcripts and truncated genes all contained MAP3K8 exons 1-8 but lacked the autoinhibitory final exon. Data mining of RNA-Seq from the Cancer Genome Atlas (TCGA) uncovered analogous MAP3K8 rearrangements in 1.5% of adult melanomas. Thus, MAP3K8 rearrangements-uncovered by comprehensive clinical sequencing of a single case-are the most common genetic event in spitzoid melanoma, are present in adult melanomas and could be amenable to MEK inhibition.
Collapse
|
40
|
Ghose R. Nature of the Pre-Chemistry Ensemble in Mitogen-Activated Protein Kinases. J Mol Biol 2018; 431:145-157. [PMID: 30562484 DOI: 10.1016/j.jmb.2018.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/09/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Abstract
In spite of the availability of a significant amount of structural detail on docking interactions involving mitogen-activated protein kinases (MAPKs) and their substrates, the mechanism by which the disordered phospho-acceptor on the substrate transiently interacts with the kinase catalytic elements and is phosphorylated, often with high efficiency, remains poorly understood. Here, this dynamic interaction is analyzed in the context of available biophysical and biochemical data for ERK2, an archetypal MAPK. A hypothesis about the nature of the ternary complex involving a MAPK, its substrate, and ATP immediately prior to the chemical step (the pre-chemistry complex) is proposed. It is postulated that the solution ensemble (the pre-chemistry ensemble) representing the pre-chemistry complex comprises several conformations that are linked by dynamics on multiple timescales. These individual conformations possess different intrinsic abilities to proceed through the chemical step. The overall rate of chemistry is therefore related to the microscopic nature of the pre-chemistry ensemble, its constituent conformational microstates, and their intrinsic abilities to yield a phosphorylated product. While characterizing these microstates within the pre-chemistry ensemble in atomic or near-atomic detail is an extremely challenging proposition, recent developments in hybrid methodologies that employ computational approaches driven by experimental data appear to provide the most promising path forward toward achieving this goal.
Collapse
Affiliation(s)
- Ranajeet Ghose
- Department of Chemistry and Biochemistry, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA; Graduate Program in Biochemistry, The Graduate Center of CUNY, New York, NY 10016, USA; Graduate Program in Chemistry, The Graduate Center of CUNY, New York, NY 10016, USA; Graduate Program in Physics, The Graduate Center of CUNY, New York, NY 10016, USA
| |
Collapse
|
41
|
Abdrabou A, Wang Z. Post-Translational Modification and Subcellular Distribution of Rac1: An Update. Cells 2018; 7:cells7120263. [PMID: 30544910 PMCID: PMC6316090 DOI: 10.3390/cells7120263] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022] Open
Abstract
Rac1 is a small GTPase that belongs to the Rho family. The Rho family of small GTPases is a subfamily of the Ras superfamily. The Rho family of GTPases mediate a plethora of cellular effects, including regulation of cytoarchitecture, cell size, cell adhesion, cell polarity, cell motility, proliferation, apoptosis/survival, and membrane trafficking. The cycling of Rac1 between the GTP (guanosine triphosphate)- and GDP (guanosine diphosphate)-bound states is essential for effective signal flow to elicit downstream biological functions. The cycle between inactive and active forms is controlled by three classes of regulatory proteins: Guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs), and guanine-nucleotide-dissociation inhibitors (GDIs). Other modifications include RNA splicing and microRNAs; various post-translational modifications have also been shown to regulate the activity and function of Rac1. The reported post-translational modifications include lipidation, ubiquitination, phosphorylation, and adenylylation, which have all been shown to play important roles in the regulation of Rac1 and other Rho GTPases. Moreover, the Rac1 activity and function are regulated by its subcellular distribution and translocation. This review focused on the most recent progress in Rac1 research, especially in the area of post-translational modification and subcellular distribution and translocation.
Collapse
Affiliation(s)
- Abdalla Abdrabou
- Department of Medical Genetics, and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Zhixiang Wang
- Department of Medical Genetics, and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
42
|
Dóczi R, Bögre L. The Quest for MAP Kinase Substrates: Gaining Momentum. TRENDS IN PLANT SCIENCE 2018; 23:918-932. [PMID: 30143312 DOI: 10.1016/j.tplants.2018.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 06/08/2023]
Abstract
Mitogen-activated protein kinase (MAPK) pathways are versatile signaling mechanisms in all eukaryotes. Their signaling outputs are defined by the protein substrates phosphorylated by MAPKs. An expanding list of substrates has been identified by high-throughput screens and targeted approaches in plants. The majority of these are phosphorylated by MPK3/6, and a few by MPK4, which are the best-characterized plant MAPKs, participating in the regulation of numerous biological processes. The identified substrates clearly represent the functional diversity of MAPKs: they are associated with pathogen defense, abiotic stress responses, ethylene signaling, and various developmental functions. Understanding their outputs is integral to unraveling the complex regulatory mechanisms of MAPK cascades. We review here methodological approaches and provide an overview of known MAPK substrates.
Collapse
Affiliation(s)
- Róbert Dóczi
- Institute of Agriculture, Centre for Agricultural Research of the Hungarian Academy of Sciences, Brunszvik utca 2, H-2462 Martonvásár, Hungary.
| | - László Bögre
- School of Biological Sciences and Centre for Systems and Synthetic Biology, Royal Holloway, University of London, Egham TW20 0EX, UK
| |
Collapse
|
43
|
Lombard CK, Davis AL, Inukai T, Maly DJ. Allosteric Modulation of JNK Docking Site Interactions with ATP-Competitive Inhibitors. Biochemistry 2018; 57:5897-5909. [PMID: 30211540 DOI: 10.1021/acs.biochem.8b00776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The c-Jun N-terminal kinases (JNKs) play a wide variety of roles in cellular signaling processes, dictating important, and even divergent, cellular fates. These essential kinases possess docking surfaces distal to their active sites that interact with diverse binding partners, including upstream activators, downstream substrates, and protein scaffolds. Prior studies have suggested that the interactions of certain protein-binding partners with one such JNK docking surface, termed the D-recruitment site (DRS), can allosterically influence the conformational state of the ATP-binding pocket of JNKs. To further explore the allosteric relationship between the ATP-binding pockets and DRSs of JNKs, we investigated how the interactions of the scaffolding protein JIP1, as well as the upstream activators MKK4 and MKK7, are allosterically influenced by the ATP-binding site occupancy of the JNKs. We show that the affinity of the JNKs for JIP1 can be divergently modulated with ATP-competitive inhibitors, with a >50-fold difference in dissociation constant observed between the lowest- and highest-affinity JNK1-inhibitor complexes. Furthermore, we found that we could promote or attenuate phosphorylation of JNK1's activation loop by MKK4 and MKK7, by varying the ATP-binding site occupancy. Given that JIP1, MKK4, and MKK7 all interact with JNK DRSs, these results demonstrate that there is functional allostery between the ATP-binding sites and DRSs of these kinases. Furthermore, our studies suggest that ATP-competitive inhibitors can allosterically influence the intracellular binding partners of the JNKs.
Collapse
Affiliation(s)
- Chloe K Lombard
- Department of Chemistry , University of Washington , Seattle , Washington 98117 , United States
| | - Audrey L Davis
- Department of Chemistry , University of Washington , Seattle , Washington 98117 , United States
| | - Takayuki Inukai
- Medicinal Chemistry Research Laboratories , Ono Pharmaceutical Company, Ltd. , 3-1-1 Sakurai , Shimamoto, Mishima, Osaka 618-8585 , Japan
| | - Dustin J Maly
- Department of Chemistry , University of Washington , Seattle , Washington 98117 , United States.,Department of Biochemistry , University of Washington , Seattle , Washington 98117 , United States
| |
Collapse
|
44
|
Sucrose Signaling Regulates Anthocyanin Biosynthesis Through a MAPK Cascade in Arabidopsis thaliana. Genetics 2018; 210:607-619. [PMID: 30143593 DOI: 10.1534/genetics.118.301470] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/06/2018] [Indexed: 01/01/2023] Open
Abstract
Anthocyanin accumulation specifically depends on sucrose (Suc) signaling. However, the molecular basis of this process remains unknown. In this study, in vitro pull-down assays identified ETHYLENE-INSENSITIVE3 (EIN3), a component of both sugar signaling or/and metabolism. This protein interacted with YDA, and the physiological relevance of this interaction was confirmed by in planta co-immunoprecipitation, yeast two-hybrid (Y2H) assay, and bimolecular fluorescence complementation. Ethylene insensitive3-like 1 (eil1) ein3 double-mutant seedlings, but not ein3-1 seedlings, showed anthocyanin accumulation. Furthermore, ein3-1 suppressed anthocyanin accumulation in yda-1 plants. Thus, EMB71/YDA-EIN3-EIL1 may form a sugar-mediated gene cascade integral to the regulation of anthocyanin accumulation. Moreover, the EMB71/YDA-EIN3-EIL1 gene cascade module directly targeted the promoter of Transparent Testa 8 (TT8) by direct EIN3 binding. Collectively, our data inferred a molecular model where the signaling cascade of the YDA-EIN3-TT8 appeared to target TT8 via EIN3, thereby modulating Suc signaling-mediated anthocyanin accumulation.
Collapse
|
45
|
Kublanovsky M, Aharoni A, Levy D. Enhanced PKMT-substrate recognition through non active-site interactions. Biochem Biophys Res Commun 2018; 501:1029-1033. [PMID: 29778536 DOI: 10.1016/j.bbrc.2018.05.103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022]
Abstract
Protein lysine methyltransferases (PKMTs) catalyze the methylation of lysine residues on many different cellular proteins. Despite extensive biochemical and structural studies, focusing on PKMT active site-peptide interactions, little is known regarding how PKMTs recognize globular substrates. To examine whether these enzymes recognize protein substrates through interactions that take place outside of the active site, we have measured SETD6 and SETD7 activity with both protein and peptide RelA substrate. We have utilized the MTase-Glo™ methyltransferase assay to measure the activity of SETD6 and SETD7 with the different RelA substrates and calculated the Michaelis-Menten (MM) parameters. We found an up to ∼12-fold increase in KM of the PKMTs activity with RelA peptide relative to the respective full-length protein, emphasizing the significantly higher PKMT-protein interaction affinity. Examination of SETD6 and SETD7 activity toward the same RelA substrates highlight the similarity in substrate recognition for both PKMTs. Our results show that the interaction affinity of SETD6 and SETD7 with RelA is enhanced through interactions that occur outside of the active site leading to higher catalytic efficiency and specificity. These interactions can significantly vary depending on the PKMT and the specific methylation site on RelA. Overall, our results underline that PKMTs can recognize their substrates through docking interactions that occur out of the active site-peptide region for enhancing their activity and specificity in the cellular environment.
Collapse
Affiliation(s)
- Margarita Kublanovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel; The National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Amir Aharoni
- The National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel; Department of Life Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| | - Dan Levy
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel; The National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| |
Collapse
|
46
|
Petrides A, Vinnicombe G. Enzyme sequestration by the substrate: An analysis in the deterministic and stochastic domains. PLoS Comput Biol 2018; 14:e1006107. [PMID: 29771922 PMCID: PMC5976211 DOI: 10.1371/journal.pcbi.1006107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 05/30/2018] [Accepted: 03/26/2018] [Indexed: 11/19/2022] Open
Abstract
This paper is concerned with the potential multistability of protein concentrations in the cell. That is, situations where one, or a family of, proteins may sit at one of two or more different steady state concentrations in otherwise identical cells, and in spite of them being in the same environment. For models of multisite protein phosphorylation for example, in the presence of excess substrate, it has been shown that the achievable number of stable steady states can increase linearly with the number of phosphosites available. In this paper, we analyse the consequences of adding enzyme docking to these and similar models, with the resultant sequestration of phosphatase and kinase by the fully unphosphorylated and by the fully phosphorylated substrates respectively. In the large molecule numbers limit, where deterministic analysis is applicable, we prove that there are always values for these rates of sequestration which, when exceeded, limit the extent of multistability. For the models considered here, these numbers are much smaller than the affinity of the enzymes to the substrate when it is in a modifiable state. As substrate enzyme-sequestration is increased, we further prove that the number of steady states will inevitably be reduced to one. For smaller molecule numbers a stochastic analysis is more appropriate, where multistability in the large molecule numbers limit can manifest itself as multimodality of the probability distribution; the system spending periods of time in the vicinity of one mode before jumping to another. Here, we find that substrate enzyme sequestration can induce bimodality even in systems where only a single steady state can exist at large numbers. To facilitate this analysis, we develop a weakly chained diagonally dominant M-matrix formulation of the Chemical Master Equation, allowing greater insights in the way particular mechanisms, like enzyme sequestration, can shape probability distributions and therefore exhibit different behaviour across different regimes.
Collapse
Affiliation(s)
- Andreas Petrides
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Glenn Vinnicombe
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
47
|
Li J, Wang Y, Meng X, Liang H. Modulation of transcriptional activity in brain lower grade glioma by alternative splicing. PeerJ 2018; 6:e4686. [PMID: 29780667 PMCID: PMC5957051 DOI: 10.7717/peerj.4686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/10/2018] [Indexed: 01/14/2023] Open
Abstract
Proteins that modify the activity of transcription factors (TFs) are often called modulators and play a vital role in gene transcriptional regulation. Alternative splicing is a critical step of gene processing, and differentially spliced isoforms may have different functions. Alternative splicing can modulate gene function by adding or removing certain protein domains and thereby influence the activity of a protein. The objective of this study is to investigate the role of alternative splicing in modulating the transcriptional regulation in brain lower grade glioma (LGG), especially transcription factor ELK1, which is closely related to various disorders, including Alzheimer’s disease and Down syndrome. The results showed that changes in the exon inclusion ratio of proteins APP and STK16 are associated with changes in the expression correlation between ELK1 and its targets. In addition, the structural features of the two modulators are strongly associated with the pathological impact of exon inclusion. The results of our analysis suggest that alternatively spliced proteins have different functions in modifying transcription factors and can thereby induce the dysregulation of multiple genes.
Collapse
Affiliation(s)
- Jin Li
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Yang Wang
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Xianglian Meng
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Hong Liang
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| |
Collapse
|
48
|
Apazoglou K, Farley S, Gorgievski V, Belzeaux R, Lopez JP, Grenier J, Ibrahim EC, El Khoury MA, Tse YC, Mongredien R, Barbé A, de Macedo CEA, Jaworski W, Bochereau A, Orrico A, Isingrini E, Guinaudie C, Mikasova L, Louis F, Gautron S, Groc L, Massaad C, Yildirim F, Vialou V, Dumas S, Marti F, Mechawar N, Morice E, Wong TP, Caboche J, Turecki G, Giros B, Tzavara ET. Antidepressive effects of targeting ELK-1 signal transduction. Nat Med 2018; 24:591-597. [DOI: 10.1038/s41591-018-0011-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/12/2018] [Indexed: 12/28/2022]
|
49
|
Rusnak L, Fu H. Regulation of ASK1 signaling by scaffold and adaptor proteins. Adv Biol Regul 2017; 66:23-30. [PMID: 29102394 DOI: 10.1016/j.jbior.2017.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 06/07/2023]
Abstract
The mitogen-activated protein kinase (MAPK) signaling pathway is a three-tiered kinase cascade where mitogen-activated protein kinase kinase kinases (MAP3Ks) lead to the activation of mitogen-activated protein kinase kinases (MAP2K), and ultimately MAPK proteins. MAPK signaling can promote a diverse set of biological outcomes, ranging from cell death to proliferation. There are multiple mechanisms which govern MAPK output, such as the duration and strength of the signal, cellular localization to upstream and downstream binding partners, pathway crosstalk and the binding to scaffold and adaptor molecules. This review will focus on scaffold and adaptor proteins that bind to and regulate apoptosis signal-regulating kinase 1 (ASK1), a MAP3K protein with a critical role in mediating stress response pathways.
Collapse
Affiliation(s)
- Lauren Rusnak
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA 30322, USA; Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, USA.
| | - Haian Fu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA 30322, USA; Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
50
|
Lu C, Liu X, Zhang CS, Gong H, Wu JW, Wang ZX. Structural and Dynamic Insights into the Mechanism of Allosteric Signal Transmission in ERK2-Mediated MKP3 Activation. Biochemistry 2017; 56:6165-6175. [PMID: 29077400 DOI: 10.1021/acs.biochem.7b00827] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mitogen-activated protein kinases (MAPKs) are key components of cellular signal transduction pathways, which are down-regulated by the MAPK phosphatases (MKPs). Catalytic activity of the MKPs is controlled both by their ability to recognize selective MAPKs and by allosteric activation upon binding to MAPK substrates. Here, we use a combination of experimental and computational techniques to elucidate the molecular mechanism for the ERK2-induced MKP3 activation. Mutational and kinetic study shows that the 334FNFM337 motif in the MKP3 catalytic domain is essential for MKP3-mediated ERK2 inactivation and is responsible for ERK2-mediated MKP3 activation. The long-term molecular dynamics (MD) simulations further reveal a complete dynamic process in which the catalytic domain of MKP3 gradually changes to a conformation that resembles an active MKP catalytic domain over the time scale of the simulation, providing a direct time-dependent observation of allosteric signal transmission in ERK2-induced MKP3 activation.
Collapse
Affiliation(s)
- Chang Lu
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University , Beijing 100084, PR China
| | - Xin Liu
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University , Beijing 100084, PR China
| | - Chen-Song Zhang
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University , Xiamen, Fujian 361005, PR China
| | - Haipeng Gong
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University , Beijing 100084, PR China
| | - Jia-Wei Wu
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University , Beijing 100084, PR China
| | - Zhi-Xin Wang
- Key Laboratory of Ministry of Education for Protein Science, School of Life Sciences, Tsinghua University , Beijing 100084, PR China
| |
Collapse
|