1
|
Haroon, Khan Z, Javaid W, Xing LX. Anthocyanin-Binding Affinity and Non-Covalent Interactions with IIS-Pathway-Related Protein Through Molecular Docking. Curr Issues Mol Biol 2025; 47:87. [PMID: 39996808 PMCID: PMC11854422 DOI: 10.3390/cimb47020087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Anthocyanins compounds, including cyanidin, malvidin, pelargonidin, peonidin, and petunidin, have demonstrated remarkable anti-aging and insulin-sensitizing properties through their interactions with proteins associated with the insulin/insulin-like growth factor signaling (IIS) pathway in Reticulitermes chinensis, employing advanced molecular docking techniques to elucidate strong binding affinities between specific anthocyanins and key proteins such as Pdk1, EIF4E, and Tsc2 in R. chinensis, suggesting a potential mechanism for their anti-aging effects. These findings not only provide critical insights into the therapeutic potential of anthocyanins for mitigating insulin resistance and promoting longevity, but also highlight the efficacy of in silico molecular docking as a predictive tool for small-molecule-protein interactions. Our research opens new avenues for the development of innovative therapeutic strategies targeting age-related diseases. However, further investigations, including a comprehensive chromosomal analysis and in vivo studies, are essential in order to fully elucidate the molecular mechanism underlying these interactions and their physiological implications. The detailed characterization of anthocyanin-binding affinities and their interactions with key regulatory genes presents exciting opportunities for advancement in molecular medicine, pharmacology, and the development of novel nutraceuticals.
Collapse
Affiliation(s)
- Haroon
- College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, Northwest University, Xi’an 710069, China;
| | - Zahid Khan
- College of Life Sciences, Northwest University, Xi’an 710069, China;
| | - Wasim Javaid
- College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, China
| | - Lian-Xi Xing
- College of Life Sciences, Northwest University, Xi’an 710069, China;
| |
Collapse
|
2
|
Alkafaas SS, Khedr SA, ElKafas SS, Hafez W, Loutfy SA, Sakran M, Janković N. Targeting JNK kinase inhibitors via molecular docking: A promising strategy to address tumorigenesis and drug resistance. Bioorg Chem 2024; 153:107776. [PMID: 39276490 DOI: 10.1016/j.bioorg.2024.107776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024]
Abstract
Among members of the mitogen-activated protein kinase (MAPK) family, c-Jun N-terminal kinases (JNKs) are vital for cellular responses to stress, inflammation, and apoptosis. Recent advances have highlighted their important implications in cancer biology, where dysregulated JNK signalling plays a role in the growth, progression, and metastasis of tumors. The present understanding of JNK kinase and its function in the etiology of cancer is summarized in this review. By modifying a number of downstream targets, such as transcription factors, apoptotic regulators, and cell cycle proteins, JNKs exert diverse effects on cancer cells. Apoptosis avoidance, cell survival, and proliferation are all promoted by abnormal JNK activation in many types of cancer, which leads to tumor growth and resistance to treatment. JNKs also affect the tumour microenvironment by controlling the generation of inflammatory cytokines, angiogenesis, and immune cell activity. However, challenges remain in deciphering the context-specific roles of JNK isoforms and their intricate crosstalk with other signalling pathways within the complex tumor environment. Further research is warranted to delineate the precise mechanisms underlying JNK-mediated tumorigenesis and to develop tailored therapeutic strategies targeting JNK signalling to improve cancer management. The review emphasizes the role of JNK kinases in cancer biology, as well as their potential as pharmaceutical targets for precision oncology therapy and cancer resistance. Also, this review summarizes all the available promising JNK inhibitors that are suggested to promote the responsiveness of cancer cells to cancer treatment.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, 31527, Egypt.
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Sara Samy ElKafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt; Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, Russia
| | - Wael Hafez
- NMC Royal Hospital, 16th St - Khalifa City - SE-4 - Abu Dhabi, United Arab Emirates; Department of Internal Medicine, Medical Research and Clinical Studies Institute, The National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate 12622, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Sakran
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Nenad Janković
- Institute for Information Technologies Kragujevac, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia.
| |
Collapse
|
3
|
Wu Y, Zhao Y, Guan Z, Esmaeili S, Xiao Z, Kuriakose D. JNK3 inhibitors as promising pharmaceuticals with neuroprotective properties. Cell Adh Migr 2024; 18:1-11. [PMID: 38357988 PMCID: PMC10878020 DOI: 10.1080/19336918.2024.2316576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 02/06/2024] [Indexed: 02/16/2024] Open
Abstract
The intensive study and investigation of neuroprotective therapy for central nervous system (CNS) diseases is ongoing. Due to shared mechanisms of neurodegeneration, a neuroprotective approach might offer benefits across multiple neurological disorders, despite variations in symptoms or injuries. C-Jun N-terminal Kinase 3 (JNK3) is found primarily in the CNS and is involved in physiological processes such as brain development, synapse formation, and memory formation. The potential of JNK3 as a target for pharmacological development holds promise for advancing neuroprotective therapies. Developing small molecule JNK3 inhibitors into drugs with neuroprotective qualities could facilitate neuronal restoration and self-repair. This review focuses on elucidating key neuroprotective mechanisms, exploring the interplay between neurodegenerative diseases and neuroprotection, and discussing advancements in JNK3 inhibitor drug development.
Collapse
Affiliation(s)
- Yibeini Wu
- Department of Anatomy and Developmental biology, Monash University, Clayton, Vic, Australia
| | - Yiling Zhao
- Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Ziman Guan
- Department of Anatomy and Developmental biology, Monash University, Clayton, Vic, Australia
| | - Sajjad Esmaeili
- Department of Anatomy and Developmental biology, Monash University, Clayton, Vic, Australia
| | - Zhicheng Xiao
- Department of Anatomy and Developmental biology, Monash University, Clayton, Vic, Australia
- Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Diji Kuriakose
- Department of Anatomy and Developmental biology, Monash University, Clayton, Vic, Australia
| |
Collapse
|
4
|
Devi B, Jangid K, Kumar N, Kumar V, Kumar V. Identification of potential JNK3 inhibitors through virtual screening, molecular docking and molecular dynamics simulation as therapeutics for Alzheimer's disease. Mol Divers 2024; 28:4361-4380. [PMID: 38573427 DOI: 10.1007/s11030-024-10820-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/30/2024] [Indexed: 04/05/2024]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder and no effective drug is available for its treatment. Numerous pathological conditions are believed to be responsible for the initiation and development of AD including c-Jun N-terminal kinases (JNKs). The JNKs are one of the enzymes from the mitogen-activated protein kinase (MAPK) family that controls the phosphorylation of various transcription factors on serine and threonine residues, and hold significant responsibilities in tasks like gene expression, cell proliferation, differentiation, and apoptosis. Since, JNK3 is primarily expressed in the brain hence its increased levels in the brain are associated with the AD pathology promoting neurofibrillary tangles, senile plaques, neuroinflammation, and nerve cell apoptosis. The current research work is focused on the development of novel JNK inhibitors as therapeutics for AD employing a structure-based virtual screening (SBVS) approach. The ZINC database (14634052 compounds) was investigated after employing pan assay interference (PAINs), drug-likeness, and diversity picking filter to distinguish molecules interacting with JNK3 by following three docking precision criteria: High Throughput Virtual Screening (HTVS), Standard Precision (SP), and Extra Precision (XP) & MMGBSA. Five lead molecules showed a better docking score in the range of -13.091 to -14.051 kcal/mol better than the reference compound (- 11.828 kcal/mol). The lead compounds displayed acceptable pharmacokinetic properties and were subjected to molecular dynamic simulations of 100 ns and binding free energy calculations. All the lead molecules showed stable RMSD and hydrogen bond interactions throughout the trajectory. The ∆GMM/PBSA_total score for the lead compounds ZINC220382956, ZINC147071339, ZINC207081127, ZINC205151456, ZINC1228819126, and CC-930 was calculated and found to be - 31.39, - 42.8, - 37.04, - 39.01, - 36.5, - 34.16 kcal/mol, respectively. Thus, it was concluded that the lead molecules identified in these studies have the potential to be explored as potent JNK3 inhibitors.
Collapse
Affiliation(s)
- Bharti Devi
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Kailash Jangid
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry and Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Naveen Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Vinay Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Vinod Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India.
| |
Collapse
|
5
|
Liu G, Gao L, Wang Y, Xie X, Gao X, Wu X. The JNK signaling pathway in intervertebral disc degeneration. Front Cell Dev Biol 2024; 12:1423665. [PMID: 39364138 PMCID: PMC11447294 DOI: 10.3389/fcell.2024.1423665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Intervertebral disc degeneration (IDD) serves as the underlying pathology for various spinal degenerative conditions and is a primary contributor to low back pain (LBP). Recent studies have revealed a strong correlation between IDD and biological processes such as Programmed Cell Death (PCD), cellular senescence, inflammation, cell proliferation, extracellular matrix (ECM) degradation, and oxidative stress (OS). Of particular interest is the emerging evidence highlighting the significant involvement of the JNK signaling pathway in these fundamental biological processes of IDD. This paper explores the potential mechanisms through the JNK signaling pathway influences IDD in diverse ways. The objective of this article is to offer a fresh perspective and methodology for in-depth investigation into the pathogenesis of IDD by thoroughly examining the interplay between the JNK signaling pathway and IDD. Moreover, this paper summarizes the drugs and natural compounds that alleviate the progression of IDD by regulating the JNK signaling pathway. This paper aims to identify potential therapeutic targets and strategies for IDD treatment, providing valuable insights for clinical application.
Collapse
Affiliation(s)
- Ganggang Liu
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lu Gao
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuncai Wang
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinsheng Xie
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xuejiao Gao
- Otolaryngology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xingjie Wu
- Orthopaedics, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Shaalan MM, Osman EEA, Attia YM, Hammam OA, George RF, Naguib BH. Novel 3,6-Disubstituted Pyridazine Derivatives Targeting JNK1 Pathway: Scaffold Hopping and Hybridization-Based Design, Synthesis, Molecular Modeling, and In Vitro and In Vivo Anticancer Evaluation. ACS OMEGA 2024; 9:37310-37329. [PMID: 39246493 PMCID: PMC11375727 DOI: 10.1021/acsomega.4c05250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
A series of novel 3,6-disubstituted pyridazine derivatives were designed, synthesized, and biologically evaluated as preclinical anticancer candidates. Compound 9e exhibited the highest growth inhibition against most of the NCI-60 cancer cell lines. The in vivo anticancer activity of 9e was subsequently investigated at two dose levels using the Ehrlich ascites carcinoma solid tumor animal model, where a reduction in the mean tumor volume allied with necrosis induction was reported without any signs of toxicity in the treated groups. Interestingly, compound 9e was capable of downregulating c-jun N-terminal kinase-1 (JNK1) gene expression and curbing the protein levels of its phosphorylated form, in parallel with a reduction in its downstream targets, namely, c-Jun and c-Fos in tumors, along with restoring p53 activity. Furthermore, molecular docking and dynamics simulations were carried out to predict the binding mode of 9e and prove its stability in the JNK1 binding pocket.
Collapse
Affiliation(s)
- Mai M Shaalan
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, Al-Sherouk City, Cairo-Suez Desert Road, Cairo 11837, Egypt
| | - Essam Eldin A Osman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Yasmeen M Attia
- Pharmacology Department, Faculty of Pharmacy, The British University in Egypt, Al-Sherouk City, Cairo-Suez Desert Road, Cairo 11837, Egypt
| | - Olfat A Hammam
- Pathology Department, Theodor Bilharz Research Institute, Imbaba, Giza 12411, Egypt
| | - Riham F George
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Bassem H Naguib
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, Al-Sherouk City, Cairo-Suez Desert Road, Cairo 11837, Egypt
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| |
Collapse
|
7
|
Mohamed GA, El-Agamy DS, Abdallah HM, Sindi IA, Almogaddam MA, Alzain AA, Andijani YS, Ibrahim SR. Kaempferol sophoroside glucoside mitigates acetaminophen-induced hepatotoxicity: Role of Nrf2/NF-κB and JNK/ASK-1 signaling pathways. Heliyon 2024; 10:e31448. [PMID: 38813141 PMCID: PMC11133934 DOI: 10.1016/j.heliyon.2024.e31448] [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: 02/01/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
APAP (Acetaminophen)-induced hepatic injury is a major public health threat that requires continuous searching for new effective therapeutics. KSG (Kaempferol-3-sophoroside-7-glucoside) is a kaempferol derivative that was separated from plant species belonging to different genera. This study explored the protective effects of KSG on ALI (acute liver injury) caused by APAP overdose in mice and elucidated its possible mechanisms. The results showed that KSG pretreatment alleviated APAP-induced hepatic damage as it reduced hepatic pathological lesions as well as the serum parameters of liver injury. Moreover, KSG opposed APAP-associated oxidative stress and augmented hepatic antioxidants. KSG suppressed the inflammatory response as it decreased the genetic and protein expression as well as the levels of inflammatory cytokines. Meanwhile, KSG enhanced the mRNA expression and level of anti-inflammatory cytokine, IL-10 (interleukin-10). KSG repressed the activation of NF-κB (nuclear-factor kappa-B), besides it promoted the activation of Nrf2 signaling. Additionally, KSG markedly hindered the elevation of ASK-1 (apoptosis-signal regulating-kinase-1) and JNK (c-Jun-N-terminal kinase). Furthermore, KSG suppressed APAP-induced apoptosis as it decreased the level and expression of Bax (BCL2-associated X-protein), and caspase-3 concurrent with an enhancement of anti-apoptotic protein, Bcl2 in the liver. More thoroughly, Computational studies reveal indispensable binding affinities between KSG and Keap1 (Kelch-like ECH-associated protein-1), ASK1 (apoptosis signal-regulating kinase-1), and JNK1 (c-Jun N-terminal protein kinase-1) with distinctive tendencies for selective inhibition. Taken together, our data showed the hepatoprotective capacity of KSG against APAP-produced ALI via modulation of Nrf2/NF-κB and JNK/ASK-1/caspase-3 signaling. Henceforth, KSG could be a promising hepatoprotective candidate for ALI.
Collapse
Affiliation(s)
- Gamal A. Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Dina S. El-Agamy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Hossam M. Abdallah
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mohammed A. Almogaddam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, 21111, Sudan
| | - Abdulrahim A. Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, 21111, Sudan
| | - Yusra Saleh Andijani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah, 30078, Saudi Arabia
| | - Sabrin R.M. Ibrahim
- Department of Chemistry, Preparatory Year Program, Batterjee Medical College, Jeddah, 21442, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
8
|
Yan H, He L, Lv D, Yang J, Yuan Z. The Role of the Dysregulated JNK Signaling Pathway in the Pathogenesis of Human Diseases and Its Potential Therapeutic Strategies: A Comprehensive Review. Biomolecules 2024; 14:243. [PMID: 38397480 PMCID: PMC10887252 DOI: 10.3390/biom14020243] [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: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
JNK is named after c-Jun N-terminal kinase, as it is responsible for phosphorylating c-Jun. As a member of the mitogen-activated protein kinase (MAPK) family, JNK is also known as stress-activated kinase (SAPK) because it can be activated by extracellular stresses including growth factor, UV irradiation, and virus infection. Functionally, JNK regulates various cell behaviors such as cell differentiation, proliferation, survival, and metabolic reprogramming. Dysregulated JNK signaling contributes to several types of human diseases. Although the role of the JNK pathway in a single disease has been summarized in several previous publications, a comprehensive review of its role in multiple kinds of human diseases is missing. In this review, we begin by introducing the landmark discoveries, structures, tissue expression, and activation mechanisms of the JNK pathway. Next, we come to the focus of this work: a comprehensive summary of the role of the deregulated JNK pathway in multiple kinds of diseases. Beyond that, we also discuss the current strategies for targeting the JNK pathway for therapeutic intervention and summarize the application of JNK inhibitors as well as several challenges now faced. We expect that this review can provide a more comprehensive insight into the critical role of the JNK pathway in the pathogenesis of human diseases and hope that it also provides important clues for ameliorating disease conditions.
Collapse
Affiliation(s)
- Huaying Yan
- Department of Ultrasound, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (H.Y.); (L.H.)
| | - Lanfang He
- Department of Ultrasound, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (H.Y.); (L.H.)
| | - De Lv
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Jun Yang
- Cancer Center and State Key Laboratory of Biotherapy, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Zhu Yuan
- Cancer Center and State Key Laboratory of Biotherapy, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
9
|
Mohamed SK, Karthikeyan S, A Omran O, Ahsin A, Salah H, Mague JT, Al-Salahi R, El Bakri Y. Insights into the crystal structure investigation and virtual screening approach of quinoxaline derivatives as potent against c-Jun N-terminal kinases 1. J Biomol Struct Dyn 2024:1-20. [PMID: 38321917 DOI: 10.1080/07391102.2024.2305317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
Quinoxaline derivatives are an important class of heterocyclic compounds in which N replaces one or more carbon atoms of the naphthalene ring and exhibit a wide spectrum of biological activities and therapeutic applications. As a result, we were encouraged to explore a new synthetic approach to quinoxaline derivatives. In this work, we synthesized two new derivatives namely, ethyl 4-(2-ethoxy-2-oxoethyl)-3-oxo-3,4-dihydroquinoxaline-2-carboxylate (2) and 3-oxo-3,4-dihydroquinoxaline-2-carbohydrazide (3) respectively. Their structures were confirmed by single-crystal X-ray analysis. Hirshfeld surface (HS) analysis is performed to understand the nature and magnitude of intermolecular interactions in the crystal packing. Density functional theory using the wb97xd/def2-TZVP method was chosen to explore their reactivity, electronic stability and optical properties. Charge transfer (CT) and orbital energies were analyzed via natural population analysis (NPA), and frontier molecular orbital (FMO) theory. The calculated excellent static hyperpolarizability (βo) indicates nonlinear optical (NLO) properties for 2 and 3. Both compounds show potent activity against c-Jun N-terminal kinases 1 (JNK 1) based on structural activity relationship studies, further subjected to molecular docking, molecular dynamics and ADMET analysis to understand their potential as drug candidates.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shaaban K Mohamed
- Chemistry and Environmental Division, Manchester Metropolitan University, Manchester, England
| | - Subramani Karthikeyan
- Center for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology University, Chennai, India
| | - Omran A Omran
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Atazaz Ahsin
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hanan Salah
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Joel T Mague
- Department of Chemistry, Tulane University, New Orleans, LA, USA
| | - Rashad Al-Salahi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Youness El Bakri
- Department of Theoretical and Applied Chemistry, South Ural State University, Chelyabinsk, Russian Federation
| |
Collapse
|
10
|
R C, M S C, M A, K S K. Crystal structure determination, molecular docking and dynamics of arylidene cyanoacetates as potential JNK-3 inhibitors for Ischemia reperfusion injury. J Biomol Struct Dyn 2023; 41:8383-8391. [PMID: 36255171 DOI: 10.1080/07391102.2022.2134207] [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: 07/04/2022] [Accepted: 10/03/2022] [Indexed: 10/24/2022]
Abstract
Ischemia reperfusion injury is a cardiovascular condition which causes hypoxia by means of obstruction of arterial blood flow eventually leads to reduced synthesis of adenosine tri-phosphate in the mitochondria. c-Jun N-terminal kinase-3 are related to several cascade of events like apoptosis, oxidative stress and mitochondrial dysfunction which can be further related to Ischemia-reperfusion injury. The present study was aimed at determining crystal structure of the ligand by x-ray methods and to perform molecular docking and molecular dynamics studies of the arylidene cyano-acetates with c-Jun N-terminal kinase-3. The binding energy of Ethyl (2E)-2-cyano-3-(4-methoxyphenyl)prop-2-enoate is -4.462 kcal/mol and ethyl (2E)-2-cyano-3-phenylprop-2-enoate is -6.135 kcal/mol. This has created a new rational approach to drug design, where the structure of drug is designed, based on its fit to structures of receptor site, rather than basing it on analogies to other active structures. The above compounds are binding strongly with c-Jun N-terminal kinase-3 protein.[Figure: see text]Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chandan R
- Department of Physics, Jain Deemed to be University, Bengaluru, Karnataka, India
| | - Chaithanya M S
- Department of Quality Assurance, Shri Siddaganga College of Pharmacy, Tumkur, Karnataka, India
| | - Aditya M
- Department of Biotechnology, Siddaganga Institute of Technology, Tumkur, Karnataka, India
| | - Kiran K S
- Department of Engineering, Faculty of Engineering and Technology, Jain Deemed to be University, Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Juyoux P, Galdadas I, Gobbo D, von Velsen J, Pelosse M, Tully M, Vadas O, Gervasio FL, Pellegrini E, Bowler MW. Architecture of the MKK6-p38α complex defines the basis of MAPK specificity and activation. Science 2023; 381:1217-1225. [PMID: 37708276 PMCID: PMC7615176 DOI: 10.1126/science.add7859] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/09/2023] [Indexed: 09/16/2023]
Abstract
The mitogen-activated protein kinase (MAPK) p38α is a central component of signaling in inflammation and the immune response and is, therefore, an important drug target. Little is known about the molecular mechanism of its activation by double phosphorylation from MAPK kinases (MAP2Ks), because of the challenge of trapping a transient and dynamic heterokinase complex. We applied a multidisciplinary approach to generate a structural model of p38α in complex with its MAP2K, MKK6, and to understand the activation mechanism. Integrating cryo-electron microscopy with molecular dynamics simulations, hydrogen-deuterium exchange mass spectrometry, and experiments in cells, we demonstrate a dynamic, multistep phosphorylation mechanism, identify catalytically relevant interactions, and show that MAP2K-disordered amino termini determine pathway specificity. Our work captures a fundamental step of cell signaling: a kinase phosphorylating its downstream target kinase.
Collapse
Affiliation(s)
- Pauline Juyoux
- European Molecular Biology Laboratory (EMBL), Grenoble, France
| | - Ioannis Galdadas
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Dorothea Gobbo
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Jill von Velsen
- European Molecular Biology Laboratory (EMBL), Grenoble, France
| | - Martin Pelosse
- European Molecular Biology Laboratory (EMBL), Grenoble, France
| | - Mark Tully
- European Synchrotron Radiation Facility, Grenoble, France
| | - Oscar Vadas
- Protein and peptide purification platform, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Francesco Luigi Gervasio
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Department of Chemistry, University College London, London, UK
- Institute of Structural and Molecular Biology, University College London, London, UK
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | | | | |
Collapse
|
12
|
Sonidegib Suppresses Production of Inflammatory Mediators and Cell Migration in BV2 Microglial Cells and Mice Treated with Lipopolysaccharide via JNK and NF-κB Inhibition. Int J Mol Sci 2022; 23:ijms231810590. [PMID: 36142500 PMCID: PMC9503982 DOI: 10.3390/ijms231810590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022] Open
Abstract
Our structure-based virtual screening of the FDA-approved drug library has revealed that sonidegib, a smoothened antagonist clinically used to treat basal cell carcinoma, is a potential c-Jun N-terminal kinase 3 (JNK3) inhibitor. This study investigated the binding of sonidegib to JNK3 via 19F NMR and its inhibitory effect on JNK phosphorylation in BV2 cells. Pharmacological properties of sonidegib to exert anti-inflammatory and anti-migratory effects were also characterized. We found that sonidegib bound to the ATP binding site of JNK3 and inhibited JNK phosphorylation in BV2 cells, confirming our virtual screening results. Sonidegib also inhibited the phosphorylation of MKK4 and c-Jun, the upstream and downstream signals of JNK, respectively. It reduced the lipopolysaccharide (LPS)-induced production of pro-inflammatory factors, including interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), and nitric oxide (NO), and the expression of inducible NO synthase and cyclooxygenase-2. The LPS-induced cell migration was suppressed by sonidegib. Sonidegib inhibited the LPS-induced IκBα phosphorylation, thereby blocking NF-κB nuclear translocation. Consistent with these findings, orally administered sonidegib attenuated IL-6 and TNF-α levels in the brains of LPS-treated mice. Collectively, our results indicate that sonidegib suppresses inflammation and cell migration in LPS-treated BV2 cells and mice by inhibiting JNK and NF-κB signaling. Therefore, sonidegib may be implicated for drug repurposing to alleviate neuroinflammation associated with microglial activation.
Collapse
|
13
|
Schepetkin IA, Kovrizhina AR, Stankevich KS, Khlebnikov AI, Kirpotina LN, Quinn MT, Cook MJ. Design, synthesis and biological evaluation of novel O-substituted tryptanthrin oxime derivatives as c-Jun N-terminal kinase inhibitors. Front Pharmacol 2022; 13:958687. [PMID: 36172181 PMCID: PMC9510750 DOI: 10.3389/fphar.2022.958687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The c-Jun N-terminal kinase (JNK) family includes three proteins (JNK1-3) that regulate many physiological processes, including inflammatory responses, morphogenesis, cell proliferation, differentiation, survival, and cell death. Therefore, JNK represents an attractive target for therapeutic intervention. Herein, a panel of novel tryptanthrin oxime analogs were synthesized and evaluated for JNK1-3 binding (Kd) and inhibition of cellular inflammatory responses (IC50). Several compounds exhibited submicromolar JNK binding affinity, with the most potent inhibitor being 6-(acetoxyimino)indolo[2,1-b]quinazolin-12(6H)-one (1j), which demonstrated high JNK1-3 binding affinity (Kd = 340, 490, and 180 nM for JNK1, JNK2, and JNK3, respectively) and inhibited lipopolysaccharide (LPS)-induced nuclear factor-κB/activating protein 1 (NF-κB/AP-1) transcription activity in THP-1Blue cells and interleukin-6 (IL-6) production in MonoMac-6 monocytic cells (IC50 = 0.8 and 1.7 μM, respectively). Compound 1j also inhibited LPS-induced production of several other proinflammatory cytokines, including IL-1α, IL-1β, granulocyte-macrophage colony-stimulating factor (GM-CSF), monocyte chemoattractant protein-1 (MCP-1), and tumor necrosis factor (TNF) in MonoMac-6 cells. Likewise, 1j inhibited LPS-induced c-Jun phosphorylation in MonoMac-6 cells, directly confirming JNK inhibition. Molecular modeling suggested modes of binding interaction of selected compounds in the JNK3 catalytic site that were in agreement with the experimental JNK3 binding data. Our results demonstrate the potential for developing anti-inflammatory drugs based on these nitrogen-containing heterocyclic systems.
Collapse
Affiliation(s)
- Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | | | - Ksenia S. Stankevich
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, United States
| | | | - Liliya N. Kirpotina
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
- *Correspondence: Mark T. Quinn, ; Matthew J. Cook,
| | - Matthew J. Cook
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, United States
- *Correspondence: Mark T. Quinn, ; Matthew J. Cook,
| |
Collapse
|
14
|
Fragment-Based and Structural Investigation for Discovery of JNK3 Inhibitors. Pharmaceutics 2022; 14:pharmaceutics14091900. [PMID: 36145648 PMCID: PMC9501523 DOI: 10.3390/pharmaceutics14091900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/21/2022] Open
Abstract
The c-Jun N-terminal kinases (JNKs) are members of the mitogen-activated protein kinase (MAPK) family and are related to cell proliferation, gene expression, and cell death. JNK isoform 3 (JNK3) is an important therapeutic target in varieties of pathological conditions including cancers and neuronal death. There is no approved drug targeting JNKs. To discover chemical inhibitors of JNK3, virtual fragment screening, the saturation transfer difference (STD) NMR, in vitro kinase assay, and X-ray crystallography were employed. A total of 27 fragments from the virtually selected 494 compounds were identified as initial hits via STD NMR and some compounds showed the inhibition of the activity of JNK3 in vitro. The structures of JNK3 with a fragment and a potent inhibitor were determined by X-ray crystallography. The fragment and inhibitor shared a common JNK3-binding feature. The result shows that fragment screening by NMR spectroscopy is a very efficient method to screen JNK3 binders and the structure of JNK3-inhibitor complex can be used to design and develop more potent inhibitors.
Collapse
|
15
|
Schepetkin IA, Chernysheva GA, Aliev OI, Kirpotina LN, Smol’yakova VI, Osipenko AN, Plotnikov MB, Kovrizhina AR, Khlebnikov AI, Plotnikov EV, Quinn MT. Neuroprotective Effects of the Lithium Salt of a Novel JNK Inhibitor in an Animal Model of Cerebral Ischemia–Reperfusion. Biomedicines 2022; 10:biomedicines10092119. [PMID: 36140222 PMCID: PMC9495587 DOI: 10.3390/biomedicines10092119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 01/31/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs) regulate many physiological processes, including inflammatory responses, morphogenesis, cell proliferation, differentiation, survival, and cell death. Therefore, JNKs represent attractive targets for therapeutic intervention. In an effort to develop improved JNK inhibitors, we synthesized the lithium salt of 11H-indeno[1,2-b]quinoxaline-11-one oxime (IQ-1L) and evaluated its affinity for JNK and biological activity in vitro and in vivo. According to density functional theory (DFT) modeling, the Li+ ion stabilizes the six-membered ring with the 11H-indeno[1,2-b]quinoxaline-11-one (IQ-1) oximate better than Na+. Molecular docking showed that the Z isomer of the IQ-1 oximate should bind JNK1 and JNK3 better than (E)-IQ-1. Indeed, experimental analysis showed that IQ-1L exhibited higher JNK1-3 binding affinity in comparison with IQ-1S. IQ-1L also was a more effective inhibitor of lipopolysaccharide (LPS)-induced nuclear factor-κB/activating protein 1 (NF-κB/AP-1) transcriptional activity in THP-1Blue monocytes and was a potent inhibitor of proinflammatory cytokine production by MonoMac-6 monocytic cells. In addition, IQ-1L inhibited LPS-induced c-Jun phosphorylation in MonoMac-6 cells, directly confirming JNK inhibition. In a rat model of focal cerebral ischemia (FCI), intraperitoneal injections of 12 mg/kg IQ-1L led to significant neuroprotective effects, decreasing total neurological deficit scores by 28, 29, and 32% at 4, 24, and 48 h after FCI, respectively, and reducing infarct size by 52% at 48 h after FCI. The therapeutic efficacy of 12 mg/kg IQ-1L was comparable to that observed with 25 mg/kg of IQ-1S, indicating that complexation with Li+ improved efficacy of this compound. We conclude that IQ-1L is more effective than IQ-1S in treating cerebral ischemia injury and thus represents a promising anti-inflammatory compound.
Collapse
Affiliation(s)
- Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Galina A. Chernysheva
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
| | - Oleg I. Aliev
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
| | - Liliya N. Kirpotina
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Vera I. Smol’yakova
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
| | - Anton N. Osipenko
- Department of Pharmacology, Siberian State Medical University, 2 Moskovskiy tract, 634050 Tomsk, Russia
| | - Mark B. Plotnikov
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
- Radiophysical Faculty, National Research Tomsk State University, 634050 Tomsk, Russia
| | | | | | - Evgenii V. Plotnikov
- Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
- Correspondence: ; Tel.: +1-406-994-4707; Fax: +1-406-994-4303
| |
Collapse
|
16
|
Nguyen PL, Bui BP, Duong MTH, Lee K, Ahn HC, Cho J. Suppression of LPS-Induced Inflammation and Cell Migration by Azelastine through Inhibition of JNK/NF-κB Pathway in BV2 Microglial Cells. Int J Mol Sci 2021; 22:ijms22169061. [PMID: 34445767 PMCID: PMC8396433 DOI: 10.3390/ijms22169061] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs) are implicated in many neuropathological conditions, including neurodegenerative diseases. To explore potential JNK3 inhibitors from the U.S. Food and Drug Administration-approved drug library, we performed structure-based virtual screening and identified azelastine (Aze) as one of the candidates. NMR spectroscopy indicated its direct binding to the ATP-binding site of JNK3, validating our observations. Although the antihistamine effect of Aze is well documented, the involvement of the JNK pathway in its action remains to be elucidated. This study investigated the effects of Aze on lipopolysaccharide (LPS)-induced JNK phosphorylation, pro-inflammatory mediators, and cell migration in BV2 microglial cells. Aze was found to inhibit the LPS-induced phosphorylation of JNK and c-Jun. It also inhibited the LPS-induced production of pro-inflammatory mediators, including interleukin-6, tumor necrosis factor-α, and nitric oxide. Wound healing and transwell migration assays indicated that Aze attenuated LPS-induced BV2 cell migration. Furthermore, Aze inhibited LPS-induced IκB phosphorylation, thereby suppressing nuclear translocation of NF-κB. Collectively, our data demonstrate that Aze exerts anti-inflammatory and anti-migratory effects through inhibition of the JNK/NF-κB pathway in BV2 cells. Based on our findings, Aze may be a potential candidate for drug repurposing to mitigate neuroinflammation in various neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases.
Collapse
|
17
|
Huang Y, Nie XM, Zhu ZJ, Zhang X, Li BZ, Ge JC, Ren Q. A novel JNK induces innate immune response by activating the expression of antimicrobial peptides in Chinese mitten crab Eriocheir sinensis. Mol Immunol 2021; 138:76-86. [PMID: 34364075 DOI: 10.1016/j.molimm.2021.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
c-Jun NH2-terminal kinase (JNK) is a member of mitogen-activated protein kinases (MAPKs) that participates in the regulation of various physiological and pathological processes. In this study, we identified a novel JNK (EsJNK) and determined the cDNA sequence of its isoform (EsJNK-a) from the Chinese mitten crab Eriocheir sinensis. The open reading frame (ORF) of EsJNK was predicted to encode 421 peptides with a serine/threonine protein kinase, a catalytic (S_TKc) domain, and a low complexity region. The ORF of EsJNK-a was 1380 bp encoding a protein with 459 amino acids, which was 38 amino acids more than that of EsJNK. The predicted tertiary structure of EsJNK was conserved and contained 15 α-helices and 10 β-sheets. Phylogenetic tree analysis revealed that EsJNK was clustered with the JNK homologs of other crustaceans. Quantitative real-time PCR assays showed that EsJNK was expressed in all the tissues examined, but it was relatively higher in hemocytes, muscles, and intestines. The expression of EsJNK mRNA in the hemocytes was upregulated by lipopolysaccharides and peptidoglycans, as well as by Staphylococcus aureus or Vibrio parahaemolyticus challenge. Functionally, after silencing EsJNK by siRNA in crabs, the expression levels of two antimicrobial peptides (AMPs), namely, anti-lipopolysaccharide factor and crustin, were significantly inhibited. The purified recombinant EsJNK protein with His-tag accelerated the elimination of the aforementioned bacteria in vivo. However, knockdown of EsJNK had an opposite effect. These findings suggested that EsJNK might be involved in the antibacterial immune defense of crabs by regulating the transcription of AMPs.
Collapse
Affiliation(s)
- Ying Huang
- College of Oceanography, Hohai University, 1 Xikang Road, Nanjing, 210098, China
| | - Xi-Mei Nie
- College of Marine Science and Engineering, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, China
| | - Zheng-Jie Zhu
- Nanjing University Ecology Research Institute of Changshu (NJUecoRICH), Changshu, 215500, China
| | - Xing Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, China
| | - Bing-Zhi Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, China
| | - Jia-Chun Ge
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, 210017, China.
| | - Qian Ren
- College of Marine Science and Engineering, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, China.
| |
Collapse
|
18
|
Garg R, Kumariya S, Katekar R, Verma S, Goand UK, Gayen JR. JNK signaling pathway in metabolic disorders: An emerging therapeutic target. Eur J Pharmacol 2021; 901:174079. [PMID: 33812885 DOI: 10.1016/j.ejphar.2021.174079] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023]
Abstract
Metabolic Syndrome is a multifactorial disease associated with increased risk of cardiovascular disorders, type 2 diabetes mellitus, fatty liver disease, etc. Various stress stimuli such as reactive oxygen species, endoplasmic reticulum stress, mitochondrial dysfunction, increased cytokines, or free fatty acids are known to aggravate progressive development of hyperglycemia and hyperlipidemia. Although the exact mechanism contributing to altered metabolism is unclear. Evidence suggests stress kinase role to be a crucial one in metabolic syndrome. Stress kinase, c-jun N-terminal kinase activation (JNK) is involved in various metabolic manifestations including obesity, insulin resistance, fatty liver disease as well as cardiometabolic disorders. It emerged as a foremost mediator in regulating metabolism in the liver, skeletal muscle, adipose tissue as well as pancreatic β cells. It has three isoforms each having a unique and tissue-specific role in altered metabolism. Current findings based on genetic manipulation or chemical inhibition studies identified JNK isoforms to play a central role in the regulation of whole-body metabolism, suggesting it to be a novel therapeutic target. Hence, it is imperative to elucidate its role in metabolic syndrome onset and progression. The purpose of this review is to elucidate in vitro and in vivo implications of JNK signaling along with the therapeutic strategy to inhibit specific isoform. Since metabolic syndrome is an array of diseases and complex pathway, carefully examining each tissue will be important for specific treatment strategies.
Collapse
Affiliation(s)
- Richa Garg
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanjana Kumariya
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Roshan Katekar
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Saurabh Verma
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Umesh K Goand
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
19
|
Hepp Rehfeldt SC, Majolo F, Goettert MI, Laufer S. c-Jun N-Terminal Kinase Inhibitors as Potential Leads for New Therapeutics for Alzheimer's Diseases. Int J Mol Sci 2020; 21:E9677. [PMID: 33352989 PMCID: PMC7765872 DOI: 10.3390/ijms21249677] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's Disease (AD) is becoming more prevalent as the population lives longer. For individuals over 60 years of age, the prevalence of AD is estimated at 40.19% across the world. Regarding the cognitive decline caused by the disease, mitogen-activated protein kinases (MAPK) pathways such as the c-Jun N-terminal kinase (JNK) pathway are involved in the progressive loss of neurons and synapses, brain atrophy, and augmentation of the brain ventricles, being activated by synaptic dysfunction, oxidative stress, and excitotoxicity. Nowadays, AD symptoms are manageable, but the disease itself remains incurable, thus the inhibition of JNK3 has been explored as a possible therapeutic target, considering that JNK is best known for its involvement in propagating pro-apoptotic signals. This review aims to present biological aspects of JNK, focusing on JNK3 and how it relates to AD. It was also explored the recent development of inhibitors that could be used in AD treatment since several drugs/compounds in phase III clinical trials failed. General aspects of the MAPK family, therapeutic targets, and experimental treatment in models are described and discussed throughout this review.
Collapse
Affiliation(s)
- Stephanie Cristine Hepp Rehfeldt
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
| | - Fernanda Majolo
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre CEP 90619-900, Rio Grande do Sul, Brazil
| | - Márcia Inês Goettert
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
| | - Stefan Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Faculty of Sciences, University of Tuebingen, D-72076 Tuebingen, Germany
| |
Collapse
|
20
|
Musi CA, Agrò G, Santarella F, Iervasi E, Borsello T. JNK3 as Therapeutic Target and Biomarker in Neurodegenerative and Neurodevelopmental Brain Diseases. Cells 2020; 9:cells9102190. [PMID: 32998477 PMCID: PMC7600688 DOI: 10.3390/cells9102190] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 01/01/2023] Open
Abstract
The c-Jun N-terminal kinase 3 (JNK3) is the JNK isoform mainly expressed in the brain. It is the most responsive to many stress stimuli in the central nervous system from ischemia to Aβ oligomers toxicity. JNK3 activity is spatial and temporal organized by its scaffold protein, in particular JIP-1 and β-arrestin-2, which play a crucial role in regulating different cellular functions in different cellular districts. Extensive evidence has highlighted the possibility of exploiting these adaptors to interfere with JNK3 signaling in order to block its action. JNK plays a key role in the first neurodegenerative event, the perturbation of physiological synapse structure and function, known as synaptic dysfunction. Importantly, this is a common mechanism in many different brain pathologies. Synaptic dysfunction and spine loss have been reported to be pharmacologically reversible, opening new therapeutic directions in brain diseases. Being JNK3-detectable at the peripheral level, it could be used as a disease biomarker with the ultimate aim of allowing an early diagnosis of neurodegenerative and neurodevelopment diseases in a still prodromal phase.
Collapse
Affiliation(s)
- Clara Alice Musi
- Department of Pharmacological and Biomolecular Sciences, Milan University, 20133 Milan, Italy;
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
| | - Graziella Agrò
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
| | - Francesco Santarella
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
| | - Erika Iervasi
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
- Department of Experimental Medicine, University of Genoa, Via De Toni 14, 16132 Genoa, Italy
| | - Tiziana Borsello
- Department of Pharmacological and Biomolecular Sciences, Milan University, 20133 Milan, Italy;
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
- Correspondence: or ; Tel.: +39-023-901-4469; Fax: +39-023-900-1916
| |
Collapse
|
21
|
Sanna MD, Manassero G, Vercelli A, Herdegen T, Galeotti N. The isoform-specific functions of the c-Jun N-terminal kinase (JNK) in a mouse model of antiretroviral-induced painful peripheral neuropathy. Eur J Pharmacol 2020; 880:173161. [DOI: 10.1016/j.ejphar.2020.173161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/30/2020] [Accepted: 04/28/2020] [Indexed: 01/26/2023]
|
22
|
Nakano R, Nakayama T, Sugiya H. Biological Properties of JNK3 and Its Function in Neurons, Astrocytes, Pancreatic β-Cells and Cardiovascular Cells. Cells 2020; 9:cells9081802. [PMID: 32751228 PMCID: PMC7464089 DOI: 10.3390/cells9081802] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022] Open
Abstract
JNK is a protein kinase, which induces transactivation of c-jun. The three isoforms of JNK, JNK1, JNK2, and JNK3, are encoded by three distinct genes. JNK1 and JNK2 are expressed ubiquitously throughout the body. By contrast, the expression of JNK3 is limited and observed mainly in the brain, heart, and testes. Concerning the biological properties of JNKs, the contribution of upstream regulators and scaffold proteins plays an important role in the activation of JNKs. Since JNK signaling has been described as a form of stress-response signaling, the contribution of JNK3 to pathophysiological events, such as stress response or cell death including apoptosis, has been well studied. However, JNK3 also regulates the physiological functions of neurons and non-neuronal cells, such as development, regeneration, and differentiation/reprogramming. In this review, we shed light on the physiological functions of JNK3. In addition, we summarize recent advances in the knowledge regarding interactions between JNK3 and cellular reprogramming.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- Laboratory of Veterinary Radiology, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa 252-0880, Japan; (T.N.); (H.S.)
- Correspondence:
| | - Tomohiro Nakayama
- Laboratory of Veterinary Radiology, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa 252-0880, Japan; (T.N.); (H.S.)
| | - Hiroshi Sugiya
- Laboratory of Veterinary Radiology, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa 252-0880, Japan; (T.N.); (H.S.)
| |
Collapse
|
23
|
Duong MTH, Lee JH, Ahn HC. C-Jun N-terminal kinase inhibitors: Structural insight into kinase-inhibitor complexes. Comput Struct Biotechnol J 2020; 18:1440-1457. [PMID: 32637042 PMCID: PMC7327381 DOI: 10.1016/j.csbj.2020.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/07/2020] [Accepted: 06/07/2020] [Indexed: 12/12/2022] Open
Abstract
The activation of c-Jun N-terminal kinases (JNKs) plays an important role in physiological processes including neuronal function, immune activity, and development, and thus, JNKs have been a therapeutic target for various diseases such as neurodegenerative diseases, inflammation, and cancer. Efforts to develop JNK-specific inhibitors have been ongoing for several decades. In this process, the structures of JNK in complex with various inhibitors have contributed greatly to the design of novel compounds and to the elucidation of structure-activity relationships. Almost 100 JNK structures with various compounds have been determined. Here we summarize the information gained from these structures and classify the inhibitors into several groups based on the binding mode. These groups include inhibitors in the open conformation and closed conformation of the gatekeeper residue, non-ATP site binders, peptides, covalent inhibitors, and type II kinase inhibitors. Through this work, deep insight into the interaction of inhibitors with JNKs can be gained and this will be helpful for developing novel, potent, and selective inhibitors.
Collapse
Affiliation(s)
- Men Thi Hoai Duong
- Department of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, South Korea
| | - Joon-Hwa Lee
- Department of Chemistry and RINS, Gyeongsang National University, Jinju, Gyeongnam 52828, South Korea
| | - Hee-Chul Ahn
- Department of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, South Korea
| |
Collapse
|
24
|
Rajan RK, Ramanathan M. Identification and neuroprotective evaluation of a potential c-Jun N-terminal kinase 3 inhibitor through structure-based virtual screening and in-vitro assay. J Comput Aided Mol Des 2020; 34:671-682. [PMID: 32040807 DOI: 10.1007/s10822-020-00297-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/29/2020] [Indexed: 02/07/2023]
Abstract
The c-Jun N-terminal kinase 3 (JNK3) signaling cascade is activated during cerebral ischemia leading to neuronal damage. The present study was carried out to identify and evaluate novel JNK3 inhibitors using in-silico and in-vitro approach. A total of 380 JNK3 inhibitors belonging to different organic groups was collected from the previously reported literature. These molecules were used to generate a pharmacophore model. This model was used to screen a chemical database (SPECS) to identify newer molecules with similar chemical features. The top 1000 hits molecules were then docked against the JNK3 enzyme coordinate following GLIDE rigid receptor docking (RRD) protocol. Best posed molecules of RRD were used during induced-fit docking (IFD), allowing receptor flexibility. Other computational predictions such as binding free energy, electronic configuration and ADME/tox were also calculated. Inferences from the best pharmacophore model suggested that, in order to have specific JNK3 inhibitory activity, the molecules must possess one H-bond donor, two hydrophobic and two ring features. Docking studies suggested that the main interaction between lead molecules and JNK3 enzyme consisted of hydrogen bond interaction with methionine 149 of the hinge region. It was also observed that the molecule with better MM-GBSA dG binding free energy, had greater correlation with JNK3 inhibition. Lead molecule (AJ-292-42151532) with the highest binding free energy (dG = 106.8 Kcal/mol) showed better efficacy than the SP600125 (reference JNK3 inhibitor) during cell-free JNK3 kinase assay (IC50 = 58.17 nM) and cell-based neuroprotective assay (EC50 = 7.5 µM).
Collapse
Affiliation(s)
- Ravi Kumar Rajan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamilnadu, India
| | - M Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamilnadu, India.
| |
Collapse
|
25
|
Malone T, Schäfer L, Simon N, Heavey S, Cuffe S, Finn S, Moore G, Gately K. Current perspectives on targeting PIM kinases to overcome mechanisms of drug resistance and immune evasion in cancer. Pharmacol Ther 2019; 207:107454. [PMID: 31836451 DOI: 10.1016/j.pharmthera.2019.107454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/03/2019] [Indexed: 12/22/2022]
Abstract
PIM kinases are a class of serine/threonine kinases that play a role in several of the hallmarks of cancer including cell cycle progression, metabolism, inflammation and immune evasion. Their constitutively active nature and unique catalytic structure has led them to be an attractive anticancer target through the use of small molecule inhibitors. This review highlights the enhanced activity of PIM kinases in cancer that can be driven by hypoxia in the tumour microenvironment and the important role that aberrant PIM kinase activity plays in resistance mechanisms to chemotherapy, radiotherapy, anti-angiogenic therapies and targeted therapies. We highlight an interaction of PIM kinases with numerous major oncogenic players, including but not limited to, stabilisation of p53, synergism with c-Myc, and notable parallel signalling with PI3K/Akt. We provide a comprehensive overview of PIM kinase's role as an escape mechanism to targeted therapies including PI3K/mTOR inhibitors, MET inhibitors, anti-HER2/EGFR treatments and the immunosuppressant rapamycin, providing a rationale for co-targeting treatment strategies for a more durable patient response. The current status of PIM kinase inhibitors and their use as a combination therapy with other targeted agents, in addition to the development of novel multi-molecularly targeted single therapeutic agents containing a PIM kinase targeting moiety are discussed.
Collapse
Affiliation(s)
- Tom Malone
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Lea Schäfer
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Nathalie Simon
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Sinead Cuffe
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Stephen Finn
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Gillian Moore
- School of Pharmacy and Biomolecular Sciences, RCSI, Dublin, Ireland
| | - Kathy Gately
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
26
|
Tuffaha GO, Hatmal MM, Taha MO. Discovery of new JNK3 inhibitory chemotypes via QSAR-Guided selection of docking-based pharmacophores and comparison with other structure-based pharmacophore modeling methods. J Mol Graph Model 2019; 91:30-51. [PMID: 31158642 DOI: 10.1016/j.jmgm.2019.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022]
Abstract
The kinase c-Jun N-terminal Kinase 3 (JNK3) plays an important role in neurodegenerative diseases. JNK3 inhibitors have shown promising results in treating Alzheimer's and Parkinson's diseases. This prompted us to model this interesting target via three established structure-based computational workflows; namely, docking-based Comparative Intermolecular Contacts Analysis (db-CICA), pharmacophore modeling via molecular-dynamics based Ligand-Receptor Contact Analysis (md-LRCA), and QSAR-guided selection of crystallographic pharmacophores. Moreover, we compared the performances of resulting pharmacophores against binding models generated via a newly introduced technique, namely, QSAR-guided selection of docking-based pharmacophores. The resulting pharmacophores were validated by receiver operating characteristic (ROC) curve analysis and used as virtual search queries to screen the National Cancer Institute (NCI) database for promising anti-JNK3 hits of novel chemotypes. Eleven nanomolar and low micromolar hits were identified, three of which were captured by QSAR-guided docking-based pharmacophores.
Collapse
Affiliation(s)
- Ghada Omar Tuffaha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Mutasem O Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan.
| |
Collapse
|
27
|
Park JY, Qu CX, Li RR, Yang F, Yu X, Tian ZM, Shen YM, Cai BY, Yun Y, Sun JP, Chung KY. Structural Mechanism of the Arrestin-3/JNK3 Interaction. Structure 2019; 27:1162-1170.e3. [PMID: 31080119 DOI: 10.1016/j.str.2019.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/25/2019] [Accepted: 04/03/2019] [Indexed: 01/14/2023]
Abstract
Arrestins, in addition to desensitizing GPCR-induced G protein activation, also mediate G protein-independent signaling by interacting with various signaling proteins. Among these, arrestins regulate MAPK signal transduction by scaffolding mitogen-activated protein kinase (MAPK) signaling components such as MAPKKK, MAPKK, and MAPK. In this study, we investigated the binding mode and interfaces between arrestin-3 and JNK3 using hydrogen/deuterium exchange mass spectrometry, 19F-NMR, and tryptophan-induced Atto 655 fluorescence-quenching techniques. Results suggested that the β1 strand of arrestin-3 is the major and potentially only interaction site with JNK3. The results also suggested that C-lobe regions near the activation loop of JNK3 form the potential binding interface, which is variable depending on the ATP binding status. Because the β1 strand of arrestin-3 is buried by the C-terminal strand in its basal state, C-terminal truncation (i.e., pre-activation) of arrestin-3 facilitates the arrestin-3/JNK3 interaction.
Collapse
Affiliation(s)
- Ji Young Park
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Chang-Xiu Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Rui-Rui Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Fan Yang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Science, Shandong University, Jinan 250012, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Zhao-Mei Tian
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Yue-Mao Shen
- Key Laboratory of Chemical Biology, (Ministry of Education), School of Pharmaceutical Science, Shandong University, Jinan, Shandong 250012, China
| | - Bo-Yang Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Youngjoo Yun
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China.
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea.
| |
Collapse
|
28
|
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
|
29
|
Mishra P, Günther S. New insights into the structural dynamics of the kinase JNK3. Sci Rep 2018; 8:9435. [PMID: 29930333 PMCID: PMC6013471 DOI: 10.1038/s41598-018-27867-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
In this work, we study the dynamics and the energetics of the all-atom structure of a neuronal-specific serine/threonine kinase c-Jun N-terminal kinase 3 (JNK3) in three states: unphosphorylated, phosphorylated, and ATP-bound phosphorylated. A series of 2 µs atomistic simulations followed by a conformational landscape mapping and a principal component analysis supports the mechanistic understanding of the JNK3 inactivation/activation process and also indicates key structural intermediates. Our analysis reveals that the unphosphorylated JNK3 undergoes the ‘open-to-closed’ movement via a two-step mechanism. Furthermore, the phosphorylation and ATP-binding allow the JNK3 kinase to attain a fully active conformation. JNK3 is a widely studied target for small-drugs used to treat a variety of neurological disorders. We believe that the mechanistic understanding of the large-conformational changes upon the activation of JNK3 will aid the development of novel targeted therapeutics.
Collapse
Affiliation(s)
- Pankaj Mishra
- Institute of Pharmaceutical Sciences, Research Group Pharmaceutical Bioinformatics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Straße 9, 79104, Freiburg, Germany
| | - Stefan Günther
- Institute of Pharmaceutical Sciences, Research Group Pharmaceutical Bioinformatics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Straße 9, 79104, Freiburg, Germany.
| |
Collapse
|
30
|
Molecular screening and analysis of novel therapeutic inhibitors against c-Jun N-terminal kinase. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Wang S, Zhang T, Yang Z, Lin J, Cai B, Ke Q, Lan W, Shi J, Wu S, Lin W. Heme oxygenase-1 protects spinal cord neurons from hydrogen peroxide-induced apoptosis via suppression of Cdc42/MLK3/MKK7/JNK3 signaling. Apoptosis 2017; 22:449-462. [PMID: 27864650 DOI: 10.1007/s10495-016-1329-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mechanisms by which oxidative stress induces spinal cord neuron death has not been completely understood. Investigation on the molecular signal pathways involved in oxidative stress-mediated neuronal death is important for development of new therapeutics for oxidative stress-associated spinal cord disorders. In current study we examined the role of heme oxygenase-1 (HO-1) in the modulation of MLK3/MKK7/JNK3 signaling, which is a pro-apoptotic pathway, after treating primary spinal cord neurons with H2O2. We found that MLK3/MKK7/JNK3 signaling was substantially activated by H2O2 in a time-dependent manner, demonstrated by increase of activating phosphorylation of MLK3, MKK7 and JNK3. H2O2 also induced expression of HO-1. Transduction of neurons with HO-1-expressing adeno-associated virus before H2O2 treatment introduced expression of exogenous HO-1 in neurons. Exogenous HO-1 reduced phosphorylation of MLK3, MKK7 and JNK3. Consistent with its inhibitory effect on MLK3/MKK7/JNK3 signaling, exogenous HO-1 decreased H2O2-induced neuronal apoptosis and necrosis. Furthermore, we found that exogenous HO-1 inhibited expression of Cdc42, which is crucial for MLK3 activation. In addition, HO-1-induced down-regulation of MLK3/MKK7/JNK3 signaling might be related to up-regulation of microRNA-137 (mir-137). A mir-137 inhibitor alleviated the inhibitory effect of HO-1 on JNK3 activation. This inhibitor also increased neuronal death even when exogenous HO-1 was expressed. Therefore, our study suggests a novel mechanism by which HO-1 exerted its neuroprotective efficacy on oxidative stress.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Tao Zhang
- Department of Orthopedic Surgery, The Second Hospital of Fuzhou Affiliated to Xiamen University, Fuzhou, 350007, China
| | - Zhen Yang
- Department of Orthopedic Surgery, The People's Hospital of Guizhou Province, Guiyang, 550002, China
| | - Jianhua Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Bin Cai
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Qingfeng Ke
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Wenbin Lan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Jinxing Shi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Shiqiang Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Wenping Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China.
| |
Collapse
|
32
|
Romano V, de Beer TAP, Schwede T. A computational protocol to evaluate the effects of protein mutants in the kinase gatekeeper position on the binding of ATP substrate analogues. BMC Res Notes 2017; 10:104. [PMID: 28219448 PMCID: PMC5319021 DOI: 10.1186/s13104-017-2428-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 02/15/2017] [Indexed: 11/11/2022] Open
Abstract
Background The determination of specific kinase substrates in vivo is challenging due to the large number of protein kinases in cells, their substrate specificity overlap, and the lack of highly specific inhibitors. In the late 90s, Shokat and coworkers developed a protein engineering-based method addressing the question of identification of substrates of protein kinases. The approach was based on the mutagenesis of the gatekeeper residue within the binding site of a protein kinase to change the co-substrate specificity from ATP to ATP analogues. One of the challenges in applying this method to other kinase systems is to identify the optimal combination of mutation in the enzyme and chemical derivative such that the ATP analogue acts as substrate for the engineered, but not the native kinase enzyme. In this study, we developed a computational protocol for estimating the effect of mutations at the gatekeeper position on the accessibility of ATP analogues within the binding site of engineered kinases. Results We tested the protocol on a dataset of tyrosine and serine/threonine protein kinases from the scientific literature where Shokat’s method was applied and experimental data were available. Our protocol correctly identified gatekeeper residues as the positions to mutate within the binding site of the studied kinase enzymes. Furthermore, the approach well reproduced the experimental data available in literature. Conclusions We have presented a computational protocol that scores how different mutations at the gatekeeper position influence the accommodation of various ATP analogues within the binding site of protein kinases. We have assessed our approach on protein kinases from the scientific literature and have verified the ability of the approach to well reproduce the available experimental data and identify suitable combinations of engineered kinases and ATP analogues.
Collapse
Affiliation(s)
- Valentina Romano
- Biozentrum, University of Basel, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Tjaart A P de Beer
- Biozentrum, University of Basel, Basel, Switzerland. .,SIB Swiss Institute of Bioinformatics, Basel, Switzerland.
| | - Torsten Schwede
- Biozentrum, University of Basel, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
33
|
Lin W, Wang S, Yang Z, Lin J, Ke Q, Lan W, Shi J, Wu S, Cai B. Heme Oxygenase-1 Inhibits Neuronal Apoptosis in Spinal Cord Injury through Down-Regulation of Cdc42-MLK3-MKK7-JNK3 Axis. J Neurotrauma 2017; 34:695-706. [PMID: 27526795 DOI: 10.1089/neu.2016.4608] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The mechanism by which spinal cord injury (SCI) induces neuronal death has not been thoroughly understood. Investigation on the molecular signal pathways involved in SCI-mediated neuronal apoptosis is important for development of new therapeutics for SCI. In the current study, we explore the role of heme oxygenase-1 (HO-1) in the modulation of mixed lineage kinase 3/mitogen-activated protein kinase kinase/cJUN N-terminal kinase 3 (MLK3/MKK7/JNK3) signaling, which is a pro-apoptotic pathway, after SCI. We found that MLK3/MKK7/JNK3 signaling was activated by SCI in a time-dependent manner, demonstrated by increase in activating phosphorylation of MLK3, MKK7, and JNK3. SCI also induced HO-1 expression. Administration of HO-1-expressing adeno-associated virus before SCI introduced expression of exogenous HO-1 in injured spinal cords. Exogenous HO-1 reduced phosphorylation of MLK3, MKK7, and JNK3. Consistent with its inhibitory effect on MLK3/MKK7/JNK3 signaling, exogenous HO-1 decreased SCI-induced neuronal apoptosis and improved neurological score. Further, we found that exogenous HO-1 inhibited expression of cell division cycle 42 (Cdc42), which is crucial for MLK3 activation. In vitro experiments indicated that Cdc42 was essential for neuronal apoptosis, while transduction of neurons with HO-1-expressing adeno-associated virus significantly reduced neuronal apoptosis to enhance neuronal survival. Therefore, our study disclosed a novel mechanism by which HO-1 exerted its neuroprotective efficacy. Our discovery might be valuable for developing a new therapeutic approach for SCI.
Collapse
Affiliation(s)
- Wenping Lin
- 1 Department of Orthopedic Surgery, the Second Affiliated Hospital, Fujian Medical University , Quanzhou, China
| | - Siyuan Wang
- 1 Department of Orthopedic Surgery, the Second Affiliated Hospital, Fujian Medical University , Quanzhou, China
| | - Zhen Yang
- 2 Department of Orthopedic Surgery, the People's Hospital of Guizhou Province , Guiyang, China
| | - Jianhua Lin
- 3 Department of Orthopedic Surgery, the First Affiliated Hospital, Fujian Medical University , Fuzhou, China
| | - Qingfeng Ke
- 1 Department of Orthopedic Surgery, the Second Affiliated Hospital, Fujian Medical University , Quanzhou, China
| | - Wenbin Lan
- 3 Department of Orthopedic Surgery, the First Affiliated Hospital, Fujian Medical University , Fuzhou, China
| | - Jinxing Shi
- 1 Department of Orthopedic Surgery, the Second Affiliated Hospital, Fujian Medical University , Quanzhou, China
| | - Shiqiang Wu
- 1 Department of Orthopedic Surgery, the Second Affiliated Hospital, Fujian Medical University , Quanzhou, China
| | - Bin Cai
- 4 Department of Neurology and Institute of Neurology, the First Affiliated Hospital, Fujian Medical University , Fuzhou, China
| |
Collapse
|
34
|
Park JY, Yun Y, Chung KY. Conformations of JNK3α splice variants analyzed by hydrogen/deuterium exchange mass spectrometry. J Struct Biol 2016; 197:271-278. [PMID: 27998708 DOI: 10.1016/j.jsb.2016.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 10/20/2022]
Abstract
c-Jun N-terminal kinases (JNKs) are members of the mitogen-activated protein kinase (MAPK) family that regulate apoptosis, inflammation, cytokine production, and metabolism. MAPKs undergo various splicing within their kinase domains. Unlike other MAPKs, JNKs have alternative splicing at the C-terminus, resulting in long and short variants. Functional or conformational effects due to the elongated C-terminal tail in the long splice variants have not been investigated nor has the conformation of the C-terminal tail been analyzed. Here, we analyzed the conformation of the elongated C-terminal tail and investigated conformational differences between long and short splice variants of JNKs using JNK3α2 and JNK3α1 as models. We adopted hydrogen/deuterium exchange mass spectrometry (HDX-MS) to analyze the conformation. HDX-MS revealed that the C-terminal tail is mostly intrinsically disordered, and that the conformation of the kinase domain of JNK3α2 is more dynamic than that of JNK3α1. The different conformation dynamics between long and short splice variants of JNK3α might affect the cellular functions of JNK3.
Collapse
Affiliation(s)
- Ji Young Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youngjoo Yun
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
35
|
Guo M, Wei J, Zhou Y, Qin Q. c-Jun N-terminal kinases 3 (JNK3) from orange-spotted grouper, Epinephelus coioides, inhibiting the replication of Singapore grouper iridovirus (SGIV) and SGIV-induced apoptosis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 65:169-181. [PMID: 27422159 DOI: 10.1016/j.dci.2016.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/10/2016] [Accepted: 06/10/2016] [Indexed: 06/06/2023]
Abstract
C-Jun N-terminal kinases (JNKs), a subgroup of serine-threonine protein kinases that activated by phosphorylation, are involve in physiological and pathophysiological processes. JNK3 is one of JNK proteins involved in JNK3 signaling transduction. In the present study, two JNK3 isoforms, Ec-JNK3 X1 and Ec-JNK3 X2, were cloned from orange-spotted grouper, Epinephelus coioides. Both Ec-JNK3 X1 and Ec-JNK3 X2 were mainly expressed in liver, gill, skin, brain and muscle of juvenile grouper. The relative expression of Ec-JNK3 X2 mRNA was much higher in muscle and gill than that of Ec-JNK3 X1. Isoform-specific immune response to challenges was revealed by the expression profiles in vivo. Immunofluorescence staining indicated that JNK3 was localized in the cytoplasm of grouper spleen (GS) cells and shown immune response to SGIV infection in vitro. Over-expressing Ec-JNK3 X1 and/or Ec-JNK3 X2 inhibited the SGIV infection and replication and the SGIV-induced apoptosis. To achieve the antiviral and anti-apoptosis activities, JNK3 promoted the activation of genes ISRE and type I IFN in the antiviral IFN signaling pathway, and inhibited the activation of transcription factors NF-κB and p53 relating to apoptosis, respectively. Ec-JNK3 X2 showed stronger activities in antivirus and anti-apoptosis than that of Ec-JNK3 X1. Our results not only define the characterization of JNK3 but also reveal new immune functions and the molecular mechanisms of JNK3 on iridoviruses infection and the virus-induced apoptosis.
Collapse
Affiliation(s)
- Minglan Guo
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China
| | - Jingguang Wei
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China; Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China
| | - Yongcan Zhou
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, PR China
| | - Qiwei Qin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, PR China; College of Marine Sciences, South China Agricultural University, Guangzhou 510642, PR China.
| |
Collapse
|
36
|
Eo Y, Han BG, Shim M, Lim JS, Phuong TNT, Hoa PP, Ahn HC. Crystal Structures of Apo- and AMP-bound Human c-Jun N-Terminal Kinase3. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yumi Eo
- Department of Pharmacy; Dongguk University-Seoul; Gyeonggi 10326 Republic of Korea
| | - Byeong Gu Han
- Department of Pharmacy; Dongguk University-Seoul; Gyeonggi 10326 Republic of Korea
| | - Myungbo Shim
- Department of Pharmacy; Dongguk University-Seoul; Gyeonggi 10326 Republic of Korea
| | - Jong Soo Lim
- Department of Pharmacy; Dongguk University-Seoul; Gyeonggi 10326 Republic of Korea
- Dong-a Socio Holdings Research Institute; Gyeonggi 17073 Republic of Korea
| | | | - Phuong Pham Hoa
- Department of Pharmacy; Dongguk University-Seoul; Gyeonggi 10326 Republic of Korea
| | - Hee-Chul Ahn
- Department of Pharmacy; Dongguk University-Seoul; Gyeonggi 10326 Republic of Korea
| |
Collapse
|
37
|
Wang B, Qin X, Wu J, Deng H, Li Y, Yang H, Chen Z, Liu G, Ren D. Analysis of crystal structure of Arabidopsis MPK6 and generation of its mutants with higher activity. Sci Rep 2016; 6:25646. [PMID: 27160427 PMCID: PMC4861982 DOI: 10.1038/srep25646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/20/2016] [Indexed: 12/02/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) cascades, which are the highly conserved signalling modules in eukaryotic organisms, have been shown to play important roles in regulating growth, development, and stress responses. The structures of various MAPKs from yeast and animal have been solved, and structure-based mutants were generated for their function analyses, however, the structures of plant MAPKs remain unsolved. Here, we report the crystal structure of Arabidopsis MPK6 at a 3.0 Å resolution. Although MPK6 is topologically similar to ERK2 and p38, the structures of the glycine-rich loop, MAPK insert, substrate binding sites, and L16 loop in MPK6 show notable differences from those of ERK2 and p38. Based on the structural comparison, we constructed MPK6 mutants and analyzed their kinase activity both in vitro and in planta. MPK6F364L and MPK6F368L mutants, in which Phe364 and Phe368 in the L16 loop were changed to Leu, respectively, acquired higher intrinsic kinase activity and retained the normal MAPKK activation property. The expression of MPK6 mutants with basal activity is sufficient to induce camalexin biosynthesis; however, to induce ethylene and leaf senescence, the expression of MPK6 mutants with higher activity is required. The results suggest that these mutants can be used to analyze the specific biological functions of MPK6.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xinghua Qin
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Juan Wu
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hongying Deng
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yuan Li
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hailian Yang
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Zhongzhou Chen
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guoqin Liu
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Dongtao Ren
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
38
|
Lange A, Günther M, Büttner FM, Zimmermann MO, Heidrich J, Hennig S, Zahn S, Schall C, Sievers-Engler A, Ansideri F, Koch P, Laemmerhofer M, Stehle T, Laufer SA, Boeckler FM. Targeting the Gatekeeper MET146 of C-Jun N-Terminal Kinase 3 Induces a Bivalent Halogen/Chalcogen Bond. J Am Chem Soc 2015; 137:14640-52. [DOI: 10.1021/jacs.5b07090] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Andreas Lange
- Molecular
Design and Pharmaceutical Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany
- Center
for Bioinformatics Tübingen (ZBIT), Eberhard Karls Universität Tübingen, Sand 1, 72076 Tübingen, Germany
| | - Marcel Günther
- Pharmaceutical
and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Felix Michael Büttner
- Interfaculty
Institute of Biochemistry, Eberhard Karls Universität Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
| | - Markus O. Zimmermann
- Molecular
Design and Pharmaceutical Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany
- Center
for Bioinformatics Tübingen (ZBIT), Eberhard Karls Universität Tübingen, Sand 1, 72076 Tübingen, Germany
| | - Johannes Heidrich
- Molecular
Design and Pharmaceutical Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany
- Center
for Bioinformatics Tübingen (ZBIT), Eberhard Karls Universität Tübingen, Sand 1, 72076 Tübingen, Germany
| | - Susanne Hennig
- Molecular
Design and Pharmaceutical Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany
| | - Stefan Zahn
- Institute
of Physical Chemistry, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring
17, 35392 Gießen, Germany
| | - Christoph Schall
- Interfaculty
Institute of Biochemistry, Eberhard Karls Universität Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
| | - Adrian Sievers-Engler
- Pharmaceutical
(Bio)Analysis, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Francesco Ansideri
- Pharmaceutical
and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Pierre Koch
- Pharmaceutical
and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Michael Laemmerhofer
- Pharmaceutical
(Bio)Analysis, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Thilo Stehle
- Interfaculty
Institute of Biochemistry, Eberhard Karls Universität Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
- Department
of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Stefan A. Laufer
- Pharmaceutical
and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Frank M. Boeckler
- Molecular
Design and Pharmaceutical Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany
- Center
for Bioinformatics Tübingen (ZBIT), Eberhard Karls Universität Tübingen, Sand 1, 72076 Tübingen, Germany
| |
Collapse
|
39
|
Chimnaronk S, Sitthiroongruang J, Srisucharitpanit K, Srisaisup M, Ketterman AJ, Boonserm P. The crystal structure of JNK from Drosophila melanogaster reveals an evolutionarily conserved topology with that of mammalian JNK proteins. BMC STRUCTURAL BIOLOGY 2015; 15:17. [PMID: 26377800 PMCID: PMC4573485 DOI: 10.1186/s12900-015-0045-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 09/07/2015] [Indexed: 11/10/2022]
Abstract
Background The c-Jun N-terminal kinases (JNKs), members of the mitogen-activated protein kinase (MAPK) family, engage in diverse cellular responses to signals produced under normal development and stress conditions. In Drosophila, only one JNK member is present, whereas ten isoforms from three JNK genes (JNK1, 2, and 3) are present in mammalian cells. To date, several mammalian JNK structures have been determined, however, there has been no report of any insect JNK structure. Results We report the first structure of JNK from Drosophila melanogaster (DJNK). The crystal structure of the unphosphorylated form of DJNK complexed with adenylyl imidodiphosphate (AMP-PNP) has been solved at 1.79 Å resolution. The fold and topology of DJNK are similar to those of mammalian JNK isoforms, demonstrating their evolutionarily conserved structures and functions. Structural comparisons of DJNK and the closely related mammalian JNKs also allow identification of putative catalytic residues, substrate-binding sites and conformational alterations upon docking interaction with Drosophila scaffold proteins. Conclusions The DJNK structure reveals common features with those of the mammalian JNK isoforms, thereby allowing the mapping of putative catalytic and substrate binding sites. Additionally, structural changes upon peptide binding could be predicted based on the comparison with the closely-related JNK3 structure in complex with pepJIP1. This is the first structure of insect JNK reported to date, and will provide a platform for future mutational studies in Drosophila to ascertain the functional role of insect JNK. Electronic supplementary material The online version of this article (doi:10.1186/s12900-015-0045-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarin Chimnaronk
- Institute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand.
| | - Jatuporn Sitthiroongruang
- Institute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand.
| | - Kanokporn Srisucharitpanit
- Faculty of Allied Health Sciences, Burapha University, Mueang District, Saen Sook, Chonburi, 20131, Thailand.
| | - Monrudee Srisaisup
- Institute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand.
| | - Albert J Ketterman
- Institute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand.
| | - Panadda Boonserm
- Institute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
40
|
Pan J, Li H, Zhang B, Xiong R, Zhang Y, Kang WY, Chen W, Zhao ZB, Chen SD. Small peptide inhibitor of JNK3 protects dopaminergic neurons from MPTP induced injury via inhibiting the ASK1-JNK3 signaling pathway. PLoS One 2015; 10:e0119204. [PMID: 25856433 PMCID: PMC4391862 DOI: 10.1371/journal.pone.0119204] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 01/27/2015] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION AND AIMS The ASK1-JNK3 signaling pathway plays a pivotal role in the pathogenesis of Parkinson's disease (PD). The specific binding of β-arrestin2 to JNK3 is essential for activation of the ASK1-JNK3 cascade, representing a potential therapeutic target for preventing dopaminergic neuronal death in PD. The aim of this study was to identify a novel strategy for the prevention of dopaminergic neuronal death in PD. METHODS Based on the specific binding of β-arrestin2 to JNK3, a 21-amino-acid fusion peptide, termed JNK3-N-Tat, was synthesized. We evaluated the ability of this peptide to inhibit the binding of β-arrestin2 to its target domain in JNK3 in vitro and in vivo. RESULTS The JNK3-N-Tat peptide inhibited activation of the ASK1-JNK3 cascade by disrupting the interaction between β-arrestin2 and JNK3. JNK3-N-Tat exerted beneficial effects through pathways downstream of JNK3 and improved mitochondrial function, resulting in attenuated MPP+/MPTP-induced damage. JNK3-N-Tat protected mesencephalic dopaminergic neurons against MPTP-induced toxicity. CONCLUSIONS JNK3-N-Tat, a JNK3-inhibitory peptide, protects dopaminergic neurons against MPP+/MPTP-induced injury by inhibiting the ASK1-JNK3 signaling pathway.
Collapse
Affiliation(s)
- Jing Pan
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Xiong
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Yan Kang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zong-Bo Zhao
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Stem Cell Biology & Laboratory of Neurodegenerative Diseases, Institute of Health Science, Shanghai Institutes of Biological Sciences (SIBS), Chinese Academy of Science (CAS) and Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
41
|
Kim MH, Lee J, Hah JM. De Novo Design and Synthesis of a γ-Turn Peptidomimetic Scaffold and Its Application as JNK3 Allosteric Ligand. Chem Asian J 2015; 10:1318-26. [DOI: 10.1002/asia.201403417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/15/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Mi-hyun Kim
- Department of Pharmacy; College of Pharmacy & Institute of Pharmaceutical Science and Technology; Hanyang University; 55 Hanyangdaehak-ro Sangnok-gu, Ansan, Kyeonggi-do 426-791 Korea
- Department of Pharmacy; College of Pharmacy & Gachon Institute of Pharmaceutical Science; Gachon University; 155 Gaetbeol-ro Yeonsu-gu, Incheon 406-840 Korea
| | - Junghun Lee
- Department of Pharmacy; College of Pharmacy & Institute of Pharmaceutical Science and Technology; Hanyang University; 55 Hanyangdaehak-ro Sangnok-gu, Ansan, Kyeonggi-do 426-791 Korea
| | - Jung-Mi Hah
- Department of Pharmacy; College of Pharmacy & Institute of Pharmaceutical Science and Technology; Hanyang University; 55 Hanyangdaehak-ro Sangnok-gu, Ansan, Kyeonggi-do 426-791 Korea
| |
Collapse
|
42
|
Co-conserved MAPK features couple D-domain docking groove to distal allosteric sites via the C-terminal flanking tail. PLoS One 2015; 10:e0119636. [PMID: 25799139 PMCID: PMC4370755 DOI: 10.1371/journal.pone.0119636] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 02/02/2015] [Indexed: 11/19/2022] Open
Abstract
Mitogen activated protein kinases (MAPKs) form a closely related family of kinases that control critical pathways associated with cell growth and survival. Although MAPKs have been extensively characterized at the biochemical, cellular, and structural level, an integrated evolutionary understanding of how MAPKs differ from other closely related protein kinases is currently lacking. Here, we perform statistical sequence comparisons of MAPKs and related protein kinases to identify sequence and structural features associated with MAPK functional divergence. We show, for the first time, that virtually all MAPK-distinguishing sequence features, including an unappreciated short insert segment in the β4-β5 loop, physically couple distal functional sites in the kinase domain to the D-domain peptide docking groove via the C-terminal flanking tail (C-tail). The coupling mediated by MAPK-specific residues confers an allosteric regulatory mechanism unique to MAPKs. In particular, the regulatory αC-helix conformation is controlled by a MAPK-conserved salt bridge interaction between an arginine in the αC-helix and an acidic residue in the C-tail. The salt-bridge interaction is modulated in unique ways in individual sub-families to achieve regulatory specificity. Our study is consistent with a model in which the C-tail co-evolved with the D-domain docking site to allosterically control MAPK activity. Our study provides testable mechanistic hypotheses for biochemical characterization of MAPK-conserved residues and new avenues for the design of allosteric MAPK inhibitors.
Collapse
|
43
|
Structural basis and biological consequences for JNK2/3 isoform selective aminopyrazoles. Sci Rep 2015; 5:8047. [PMID: 25623238 PMCID: PMC4306959 DOI: 10.1038/srep08047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/31/2014] [Indexed: 12/19/2022] Open
Abstract
Three JNK isoforms, JNK1, JNK2, and JNK3 have been reported and unique biological function has been ascribed to each. It is unknown if selective inhibition of these isoforms would confer therapeutic or safety benefit. To probe JNK isoform function we designed JNK2/3 inhibitors that have >30-fold selectivity over JNK1. Utilizing site-directed mutagenesis and x-ray crystallography we identified L144 in JNK3 as a key residue for selectivity. To test whether JNK2/3 selective inhibitors protect human dopaminergic neurons against neurotoxin-induced mitochondrial dysfunction, we monitored reactive oxygen species (ROS) generation and mitochondrial membrane potential (MMP). The results showed that JNK2/3 selective inhibitors protected against 6-hydroxydopamine-induced ROS generation and MMP depolarization. These results suggest that it was possible to develop JNK2/3 selective inhibitors and that residues in hydrophobic pocket I were responsible for selectivity. Moreover, the findings also suggest that inhibition of JNK2/3 likely contributed to protecting mitochondrial function and prevented ultimate cell death.
Collapse
|
44
|
Zheng K, Iqbal S, Hernandez P, Park H, LoGrasso PV, Feng Y. Design and synthesis of highly potent and isoform selective JNK3 inhibitors: SAR studies on aminopyrazole derivatives. J Med Chem 2014; 57:10013-30. [PMID: 25393557 PMCID: PMC4266361 DOI: 10.1021/jm501256y] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
![]()
The
c-jun N-terminal kinase 3 (JNK3) is expressed primarily in
the brain. Numerous reports have shown that inhibition of JNK3 is
a promising strategy for treatment of neurodegeneration. The optimization
of aminopyrazole-based JNK3 inhibitors with improved potency, isoform
selectivity, and pharmacological properties by structure–activity
relationship (SAR) studies utilizing biochemical and cell-based assays,
and structure-based drug design is reported. These inhibitors had
high selectivity over JNK1 and p38α, minimal cytotoxicity, potent
inhibition of 6-OHDA-induced mitochondrial membrane potential dissipation
and ROS generation, and good drug metabolism and pharmacokinetic (DMPK)
properties for iv dosing. 26n was profiled against 464
kinases and was found to be highly selective hitting only seven kinases
with >80% inhibition at 10 μM. Moreover, 26n showed
good solubility, good brain penetration, and good DMPK properties.
Finally, the crystal structure of 26k in complex with
JNK3 was solved at 1.8 Å to explore the binding mode of aminopyrazole
based JNK3 inhibitors.
Collapse
Affiliation(s)
- Ke Zheng
- Medicinal Chemistry, ‡Discovery Biology, §Crystallography/Modeling Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, Scripps Florida, The Scripps Research Institute , 130 Scripps Way, No. 2A1, Jupiter, Florida 33458, United States
| | | | | | | | | | | |
Collapse
|
45
|
Messoussi A, Feneyrolles C, Bros A, Deroide A, Daydé-Cazals B, Chevé G, Van Hijfte N, Fauvel B, Bougrin K, Yasri A. Recent progress in the design, study, and development of c-Jun N-terminal kinase inhibitors as anticancer agents. ACTA ACUST UNITED AC 2014; 21:1433-43. [PMID: 25442375 DOI: 10.1016/j.chembiol.2014.09.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/19/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022]
Abstract
The c-Jun N-terminal kinase (JNK) family, with its three members JNK1, JNK2, and JNK3, is a subfamily of mitogen-activated protein kinases. Involved in many aspects of cellular processes, JNK has been also associated with pathological states such as neurodegenerative diseases, inflammation, and cancers. In oncology, each isoform plays a distinct role depending on the context of the targeted tissue/organ, the tumor stage, and, most likely, the signaling pathway activated upstream. Consequently, the current challenge in finding new successful anti-JNK therapies is to design isoform-selective inhibitors of the JNKs. In this review, a particular focus is given to the JNK inhibitors that have been developed thus far when examining 3D structures of various JNK-inhibitor complexes. Using current data regarding structure-activity relationships and medicinal chemistry approaches, our objective is to provide a better understanding of the design and development of selective JNK inhibitors in the present and future.
Collapse
Affiliation(s)
- Abdellah Messoussi
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 34090 Montpellier, France; Laboratoire de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Université Mohammed V-Agdal, Faculté des Sciences, B.P. 1014 Rabat, Morocco
| | | | - Aurélie Bros
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 34090 Montpellier, France
| | - Arthur Deroide
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 34090 Montpellier, France
| | | | - Gwénaël Chevé
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 34090 Montpellier, France
| | | | - Bénédicte Fauvel
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 34090 Montpellier, France
| | - Khalid Bougrin
- Laboratoire de Chimie des Plantes et de Synthèse Organique et Bioorganique, URAC23, Université Mohammed V-Agdal, Faculté des Sciences, B.P. 1014 Rabat, Morocco
| | - Aziz Yasri
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 34090 Montpellier, France.
| |
Collapse
|
46
|
Owen GR, Stoychev S, Achilonu I, Dirr HW. Phosphorylation- and nucleotide-binding-induced changes to the stability and hydrogen exchange patterns of JNK1β1 provide insight into its mechanisms of activation. J Mol Biol 2014; 426:3569-89. [PMID: 25178256 DOI: 10.1016/j.jmb.2014.08.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/22/2014] [Accepted: 08/25/2014] [Indexed: 12/20/2022]
Abstract
Many studies have characterized how changes to the stability and internal motions of a protein during activation can contribute to their catalytic function, even when structural changes cannot be observed. Here, unfolding studies and hydrogen-deuterium exchange (HX) mass spectrometry were used to investigate the changes to the stability and conformation/conformational dynamics of JNK1β1 induced by phosphorylative activation. Equivalent studies were also employed to determine the effects of nucleotide binding on both inactive and active JNK1β1 using the ATP analogue, 5'-adenylyl-imidodiphosphate (AMP-PNP). JNK1β1 phosphorylation alters HX in regions involved in catalysis and substrate binding, changes that can be ascribed to functional modifications in either structure and/or backbone flexibility. Increased HX in the hinge between the N- and C-terminal domains implied that it acquires enhanced flexibility upon phosphorylation that may be a prerequisite for interdomain closure. In combination with the finding that nucleotide binding destabilizes the kinase, the patterns of solvent protection by AMP-PNP were consistent with a novel mode of nucleotide binding to the C-terminal domain of a destabilized and open domain conformation of inactive JNK1β1. Solvent protection by AMP-PNP of both N- and C-terminal domains in active JNK1β1 revealed that the domains close around nucleotide upon phosphorylation, concomitantly stabilizing the kinase. This suggests that phosphorylation activates JNK1β1 in part by increasing hinge flexibility to facilitate interdomain closure and the creation of a functional active site. By uncovering the complex interplay that occurs between nucleotide binding and phosphorylation, we present new insight into the unique mechanisms by which JNK1β1 is regulated.
Collapse
Affiliation(s)
- Gavin R Owen
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Stoyan Stoychev
- Biosciences, Council for Scientific and Industrial Research, Pretoria 0001, South Africa
| | - Ikechukwu Achilonu
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Heini W Dirr
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa.
| |
Collapse
|
47
|
Prakasam A, Ghose S, Oleinik NV, Bethard JR, Peterson YK, Krupenko NI, Krupenko SA. JNK1/2 regulate Bid by direct phosphorylation at Thr59 in response to ALDH1L1. Cell Death Dis 2014; 5:e1358. [PMID: 25077544 PMCID: PMC4123105 DOI: 10.1038/cddis.2014.316] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/06/2014] [Accepted: 06/18/2014] [Indexed: 01/07/2023]
Abstract
BH3 interacting-domain death agonist (Bid) is a BH3-only pro-apoptotic member of the Bcl-2 family of proteins. Its function in apoptosis is associated with the proteolytic cleavage to the truncated form tBid, mainly by caspase-8. tBid translocates to mitochondria and assists Bax and Bak in induction of apoptosis. c-Jun N-terminal kinase (JNK)-dependent alternative processing of Bid to jBid was also reported. We have previously shown that the folate stress enzyme 10-formyltetrahydrofolate dehydrogenase (ALDH1L1) activates JNK1 and JNK2 in cancer cells as a pro-apoptotic response. Here we report that in PC-3 prostate cancer cells, JNK1/2 phosphorylate Bid at Thr59 within the caspase cleavage site in response to ALDH1L1. In vitro, all three JNK isoforms, JNK 1–3, phosphorylated Thr59 of Bid with JNK1 being the least active. Thr59 phosphorylation protected Bid from cleavage by caspase-8, resulting in strong accumulation of the full-length protein and its translocation to mitochondria. Interestingly, although we did not observe jBid in response to ALDH1L1 in PC-3 cells, transient expression of Bid mutants lacking the caspase-8 cleavage site resulted in strong accumulation of jBid. Of note, a T59D mutant mimicking constitutive phosphorylation revealed more profound cleavage of Bid to jBid. JNK-driven Bid accumulation had a pro-apoptotic effect in our study: small interfering RNA silencing of either JNK1/2 or Bid prevented Bid phosphorylation and accumulation, and rescued ALDH1L1-expressing cells. As full-length Bid is a weaker apoptogen than tBid, we propose that the phosphorylation of Bid by JNKs, followed by the accumulation of the full-length protein, delays attainment of apoptosis, and allows the cell to evaluate the stress and make a decision regarding the response strategy. This mechanism perhaps can be modified by the alternative cleavage of phospho-T59 Bid to jBid at some conditions.
Collapse
Affiliation(s)
- A Prakasam
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - S Ghose
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - N V Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - J R Bethard
- Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Y K Peterson
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - N I Krupenko
- 1] Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA [2] Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - S A Krupenko
- 1] Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA [2] Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
48
|
Srivastava P, Yadav RS, Chandravanshi LP, Shukla RK, Dhuriya YK, Chauhan LKS, Dwivedi HN, Pant AB, Khanna VK. Unraveling the mechanism of neuroprotection of curcumin in arsenic induced cholinergic dysfunctions in rats. Toxicol Appl Pharmacol 2014; 279:428-440. [PMID: 24952339 DOI: 10.1016/j.taap.2014.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/20/2022]
Abstract
Earlier, we found that arsenic induced cholinergic deficits in rat brain could be protected by curcumin. In continuation to this, the present study is focused to unravel the molecular mechanisms associated with the protective efficacy of curcumin in arsenic induced cholinergic deficits. Exposure to arsenic (20mg/kg body weight, p.o) for 28 days in rats resulted to decrease the expression of CHRM2 receptor gene associated with mitochondrial dysfunctions as evident by decrease in the mitochondrial membrane potential, activity of mitochondrial complexes and enhanced apoptosis both in the frontal cortex and hippocampus in comparison to controls. The ultrastructural images of arsenic exposed rats, assessed by transmission electron microscope, exhibited loss of myelin sheath and distorted cristae in the mitochondria both in the frontal cortex and hippocampus as compared to controls. Simultaneous treatment with arsenic (20mg/kg body weight, p.o) and curcumin (100mg/kg body weight, p.o) for 28 days in rats was found to protect arsenic induced changes in the mitochondrial membrane potential and activity of mitochondrial complexes both in frontal cortex and hippocampus. Alterations in the expression of pro- and anti-apoptotic proteins and ultrastructural damage in the frontal cortex and hippocampus following arsenic exposure were also protected in rats simultaneously treated with arsenic and curcumin. The data of the present study reveal that curcumin could protect arsenic induced cholinergic deficits by modulating the expression of pro- and anti-apoptotic proteins in the brain. More interestingly, arsenic induced functional and ultrastructural changes in the brain mitochondria were also protected by curcumin.
Collapse
Affiliation(s)
- Pranay Srivastava
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajesh S Yadav
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India; Department of Crimnology and Forensic Science, Harisingh Gour University, Sagar 470 003, India
| | - Lalit P Chandravanshi
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajendra K Shukla
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Yogesh K Dhuriya
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Lalit K S Chauhan
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Hari N Dwivedi
- Babu Banarasi Das University, BBD City, Faizabad Road, Lucknow 227 015, India
| | - Aditiya B Pant
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Vinay K Khanna
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India.
| |
Collapse
|
49
|
|
50
|
Hari SB, Merritt EA, Maly DJ. Sequence determinants of a specific inactive protein kinase conformation. ACTA ACUST UNITED AC 2014; 20:806-15. [PMID: 23790491 DOI: 10.1016/j.chembiol.2013.05.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/18/2013] [Accepted: 05/09/2013] [Indexed: 02/07/2023]
Abstract
Only a small percentage of protein kinases have been shown to adopt a distinct inactive ATP-binding site conformation, called the Asp-Phe-Gly-out (DFG-out) conformation. Given the high degree of homology within this enzyme family, we sought to understand the basis of this disparity on a sequence level. We identified two residue positions that sensitize mitogen-activated protein kinases (MAPKs) to inhibitors that stabilize the DFG-out inactive conformation. After characterizing the structure and dynamics of an inhibitor-sensitive MAPK mutant, we demonstrated the generality of this strategy by sensitizing a kinase (apoptosis signal-regulating kinase 1) not in the MAPK family to several DFG-out stabilizing ligands, using the same residue positions. The use of specific inactive conformations may aid the study of noncatalytic roles of protein kinases, such as binding partner interactions and scaffolding effects.
Collapse
Affiliation(s)
- Sanjay B Hari
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|