1
|
Jin Y, Zhao L, Zhang Y, Chen T, Shi H, Sun H, Ding S, Chen S, Cao H, Zhang G, Li Q, Gao J, Xiao M, Sheng C. BIN1 deficiency enhances ULK3-dependent autophagic flux and reduces dendritic size in mouse hippocampal neurons. Autophagy 2025; 21:223-242. [PMID: 39171951 DOI: 10.1080/15548627.2024.2393932] [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: 07/14/2023] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
Genome-wide association studies identified variants around the BIN1 (bridging integrator 1) gene locus as prominent risk factors for late-onset Alzheimer disease. In the present study, we decreased the expression of BIN1 in mouse hippocampal neurons to investigate its neuronal function. Bin1 knockdown via RNAi reduced the dendritic arbor size in primary cultured hippocampal neurons as well as in mature Cornu Ammonis 1 excitatory neurons. The AAV-mediated Bin1 RNAi knockdown also generated a significant regional volume loss around the injection sites at the organ level, as revealed by 7-Tesla structural magnetic resonance imaging, and an impaired spatial reference memory performance in the Barnes maze test. Unexpectedly, Bin1 knockdown led to concurrent activation of both macroautophagy/autophagy and MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1). Autophagy inhibition with the lysosome inhibitor chloroquine effectively mitigated the Bin1 knockdown-induced dendritic regression. The subsequent molecular studydemonstrated that increased expression of ULK3 (unc-51 like kinase 3), which is MTOR-insensitive, supported autophagosome formation in BIN1 deficiency. Reducing ULK3 activity with SU6668, a receptor tyrosine kinase inhibitor, or decreasing neuronal ULK3 expression through AAV-mediated RNAi, significantly attenuated Bin1 knockdown-induced hippocampal volume loss and spatial memory decline. In Alzheimer disease patients, the major neuronal isoform of BIN1 is specifically reduced. Our work suggests this reduction is probably an important molecular event that increases the autophagy level, which might subsequently promote brain atrophy and cognitive impairment through reducing dendritic structures, and ULK3 is a potential interventional target for relieving these detrimental effects.Abbreviations: AV: adeno-associated virus; Aβ: amyloid-β; ACTB: actin, beta; AD: Alzheimer disease; Aduk: Another Drosophila Unc-51-like kinase; AKT1: thymoma viral proto-oncogene 1; AMPK: AMP-activated protein kinase; AP: autophagosome; BafA1: bafilomycin A1; BDNF: brain derived neurotrophic factor; BIN1: bridging integrator 1; BIN1-iso1: BIN1, isoform 1; CA1: cornu Ammonis 1; CA3: cornu Ammonis 3; CLAP: clathrin and adapter binding; CQ: chloroquine; DMEM: Dulbecco's modified Eagle medium; EGFP: enhanced green fluorescent protein; GWAS: genome-wide association study; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MRI: magnetic resonance imaging; MTOR; mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; PET: positron emission tomography; qRT-PCR: real-time quantitative reverse transcription PCR; ROS: reactive oxygen species; RPS6KB1: ribosomal protein S6 kinase B1; TFEB: transcription factor EB; ULK1: unc-51 like kinase 1; ULK3: unc-51 like kinase 3.
Collapse
Affiliation(s)
- Yuxi Jin
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lin Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yanli Zhang
- Department of Psychiatry, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Tingzhen Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huili Shi
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Huaiqing Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shixin Ding
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Sijia Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Haifeng Cao
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Guannan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chengyu Sheng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Small Molecule Inhibitors for Unc-51-like Autophagy-Activating Kinase Targeting Autophagy in Cancer. Int J Mol Sci 2023; 24:ijms24020953. [PMID: 36674464 PMCID: PMC9866249 DOI: 10.3390/ijms24020953] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Autophagy is a cellular process that removes damaged components of cells and recycles them as biochemical building blocks. Autophagy can also be induced to protect cells in response to intra- and extracellular stresses, including damage to cellular components, nutrient deprivation, hypoxia, and pathogenic invasion. Dysregulation of autophagy has been attributed to various diseases. In particular, autophagy protects cancer cells by supporting tumor cell survival and the development of drug resistance. Understanding the pathophysiological mechanisms of autophagy in cancer has stimulated the research on discovery and development of specific inhibitors targeting various stages of autophagy. In recent years, Unc-51-like autophagy-activating kinase (ULK) inhibitors have become an attractive strategy to treat cancer. This review summarizes recent discoveries and developments in small-molecule ULK inhibitors and their potential as anticancer agents. We focused on structural features, interactions with binding sites, and biological effects of these inhibitors. Overall, this review will provide guidance for using ULK inhibitors as chemical probes for autophagy in various cancers and developing improved ULK inhibitors that would enhance therapeutic benefits in the clinic.
Collapse
|
3
|
Duan T, Yang X, Kuang J, Sun W, Li J, Ge J, Zhang M, Cai X, Yu P, Yang J, Zhu X. ULK1 Depletion Protects Mice from Diethylnitrosamine-Induced Hepatocarcinogenesis by Promoting Apoptosis and Inhibiting Autophagy. J Hepatocell Carcinoma 2023; 10:315-325. [PMID: 36874251 PMCID: PMC9983443 DOI: 10.2147/jhc.s399855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/28/2023] [Indexed: 03/03/2023] Open
Abstract
Purpose The uncoordinated-51 like kinase 1 (ULK1) is an important serine/threonine protein kinase involved in autophagy, especially for the initiation stage. Previous studies have shown that ULK1 could be used as a prognostic marker in predicting poor progression-free survival and a therapeutic target for hepatocellular carcinoma (HCC) when treated with sorafenib; however, its role during hepatocarcinogenesis remains to be elucidated. Methods CCK8 and colony formation assay were used to detect cell growth ability. Western blotting was performed to determine expression level of protein. Data from public database were downloaded to analyze expression of ULK1 at mRNA level and predict survival time. RNA-seq was conducted to reveal disturbed gene profile orchestrated by ULK1 depletion. A diethylnitrosamine (DEN)-induced HCC mice model was used to uncover the role of ULK1 in hepatocarcinogenesis. Results ULK1 was up-regulated in liver cancer tissues and cell lines, and knockdown of ULK1 promoted apoptosis and suppressed proliferation of liver cancer cells. In in vivo experiments, Ulk1 depletion attenuated starvation-induced autophagy in mice liver, reduced diethylnitrosamine (DEN)-induced hepatic tumor number and size, and prevented tumor progression. Further, RNA-seq analysis revealed a close relationship between Ulk1 and immunity with significant changes in gene sets enriched in the interleukin and interferon pathways. Conclusion ULK1 deficiency prevented hepatocarcinogenesis and inhibited hepatic tumor growth, and might be a molecular target for the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Ting Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Xin Yang
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Jingyu Kuang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, 410073, People's Republic of China
| | - Wenjie Sun
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Jin Li
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Juan Ge
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Mohan Zhang
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Xiaobo Cai
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Peilin Yu
- Department of Toxicology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Jun Yang
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, Zhejiang, 311121, People's Republic of China
| | - Xinqiang Zhu
- Medical Research Center, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, People's Republic of China
| |
Collapse
|
4
|
Rizwi FA, Abubakar M, Puppala ER, Goyal A, Bhadrawamy CV, Naidu VGM, Roshan S, Tazneem B, Almalki WH, Subramaniyan V, Rawat S, Gupta G. Janus Kinase-Signal Transducer and Activator of Transcription Inhibitors for the Treatment and Management of Cancer. J Environ Pathol Toxicol Oncol 2023; 42:15-29. [PMID: 37522565 DOI: 10.1615/jenvironpatholtoxicoloncol.2023045403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
According to the World Health Organization (WHO), cancer is the second-highest cause of mortality worldwide, killing nearly 9.6 million people annually. Despite the advances in diagnosis and treatment during the last couple of decades, it remains a serious concern due to the limitations of currently available cancer management strategies. Therefore, alternative strategies are highly required to overcome these glitches. In addition, many etiological factors such as environmental and genetic factors initiate the activation of the Janus kinase (JAK)-signal transducer and activator of the transcription (STAT) pathway. This aberrant activation of the JAK-STAT pathway has been reported in various disease states, including inflammatory conditions, hematologic malignancies, and cancer. For instance, many patients with myeloproliferative neoplasms carry the acquired gain-of-function JAK2 V617F somatic mutation. This knowledge has dramatically improved our understanding of pathogenesis and has facilitated the development of therapeutics capable of suppressing the constitutive activation of the JAK-STAT pathway. Our aim is not to be expansive but to highlight emerging ideas towards preventive therapy in a modern view of JAK-STAT inhibitors. A series of agents with different specificities against different members of the JAK family of proteins is currently undergoing evaluation in clinical trials. Here we give a summary of how JAK-STAT inhibitors function and a detailed review of current clinical drugs for managing cancer as a new therapeutic approach.
Collapse
Affiliation(s)
- Fahim Anwar Rizwi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - Md Abubakar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - Eswara Rao Puppala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Ch Veera Bhadrawamy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - S Roshan
- Deccan School of Pharmacy, Hyderabad, India
| | - B Tazneem
- Deccan School of Pharmacy, Hyderabad, India
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, MONASH University, Malaysia
| | - Sushama Rawat
- Nirma University, Institute of Pharmacy, Ahmedabad, Gujarat 382481, India; School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 302017, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 302017, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
5
|
González-Rodríguez P, Cheray M, Keane L, Engskog-Vlachos P, Joseph B. ULK3-dependent activation of GLI1 promotes DNMT3A expression upon autophagy induction. Autophagy 2022; 18:2769-2780. [PMID: 35226587 PMCID: PMC9673947 DOI: 10.1080/15548627.2022.2039993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Macroautophagy/autophagy is a tightly regulated catabolic process, which contributes at baseline level to cellular homeostasis, and upon its stimulation to the adaptive cellular response to intra- and extracellular stress stimuli. Decrease of autophagy activity is occurring upon aging and thought to contribute to age-related-diseases. Recently, we uncovered, upon autophagy induction, the role of de novo DNMT3A (DNA methyltransferase 3 alpha)-mediated DNA methylation on expression of the MAP1LC3 (microtubule associated protein 1 light chain 3) proteins, core components of the autophagy pathway, which resulted in reduced baseline autophagy activity. Here, we report that serine/threonine kinase ULK3 (unc-51 like kinase 3)-dependent activation of GLI1 (GLI family zinc finger 1) contributes to the transcriptional upregulation of DNMT3A gene expression upon autophagy induction, thereby bringing additional understanding of the long-term effect of autophagy induction and a possible mechanism for its decline upon aging, pathological conditions, or in response to treatment interventions.Abbreviations: CBZ: carbamazepine; ChIP: chromatin immunoprecipitation; Clon: clonidine; DNMT3A: DNA methyltransferase 3 alpha; GLI1: GLI family zinc finger 1; GLI2: GLI family zinc finger 2; MAP1LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PLA: proximity ligation assay; RT-qPCR: quantitative reverse transcription PCR; shRNA: small hairpin RNA; siRNA: small interfering RNA; Treh: trehalose; ULK3: unc-51 like kinase 3.
Collapse
Affiliation(s)
| | - Mathilde Cheray
- Institute of Environmental MedicineToxicology Unit, Karolinska Institutet, StockholmSweden
| | - Lily Keane
- Institute of Environmental MedicineToxicology Unit, Karolinska Institutet, StockholmSweden
| | | | - Bertrand Joseph
- Institute of Environmental MedicineToxicology Unit, Karolinska Institutet, StockholmSweden,Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway,CONTACT Bertrand Joseph Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm17177, Sweden; Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Mathavan I, Liu LJ, Robinson SW, El-Sakkary N, Elatico AJJ, Gomez D, Nellas R, Owens RJ, Zuercher W, Navratilova I, Caffrey CR, Beis K. Identification of Inhibitors of the Schistosoma mansoni VKR2 Kinase Domain. ACS Med Chem Lett 2022; 13:1715-1722. [PMID: 36385939 PMCID: PMC9661718 DOI: 10.1021/acsmedchemlett.2c00248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/30/2022] [Indexed: 02/02/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by parasitic flatworms. Current treatment relies on just one partially effective drug, praziquantel (PZQ). Schistosoma mansoni Venus Kinase Receptors 1 and 2 (SmVKR1 and SmVKR2) are important for parasite growth and egg production, and are potential targets for combating schistosomiasis. VKRs consist of an extracellular Venus Flytrap Module (VFTM) linked via a transmembrane helix to a kinase domain. Here, we initiated a drug discovery effort to inhibit the activity of the SmVKR2 kinase domain (SmVKR2KD) by screening the GSK published kinase inhibitor set 2 (PKIS2). We identified several inhibitors, of which four were able to inhibit its enzymatic activity and induced phenotypic changes in ex vivo S. mansoni. Our crystal structure of the SmVKR2KD displays an active-like state that sheds light on the activation process of VKRs. Our data provide a basis for the further exploration of SmVKR2 as a possible drug target.
Collapse
Affiliation(s)
- Indran Mathavan
- Department
of Life Sciences, Imperial College London, Exhibition Road, London, South Kensington SW7 2AZ, United Kingdom
- Rutherford
Appleton Laboratory, Research Complex at
Harwell, Didcot, Oxfordshire OX11 0FA, United Kingdom
| | - Lawrence J. Liu
- Center
for Discovery and Innovation in Parasitic Diseases, Skaggs School
of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Sean W. Robinson
- Kinetic
Discovery Ltd., an Exscientia group company, The Schrödinger Building, Oxford Science
Park, Oxford OX4 4GE, United Kingdom
| | - Nelly El-Sakkary
- Center
for Discovery and Innovation in Parasitic Diseases, Skaggs School
of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Adam Jo J. Elatico
- Institute
of Chemistry, College of Science, University
of the Philippines Diliman, Quezon City, Philippines 1101
| | - Darwin Gomez
- Institute
of Chemistry, College of Science, University
of the Philippines Diliman, Quezon City, Philippines 1101
| | - Ricky Nellas
- Institute
of Chemistry, College of Science, University
of the Philippines Diliman, Quezon City, Philippines 1101
| | - Raymond J. Owens
- The Rosalind
Franklin Institute, Harwell Campus, Didcot, OX11 0QX, United Kingdom
- Division
of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United
Kingdom
| | - William Zuercher
- Structural
Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Iva Navratilova
- Kinetic
Discovery Ltd., an Exscientia group company, The Schrödinger Building, Oxford Science
Park, Oxford OX4 4GE, United Kingdom
| | - Conor R. Caffrey
- Center
for Discovery and Innovation in Parasitic Diseases, Skaggs School
of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Konstantinos Beis
- Department
of Life Sciences, Imperial College London, Exhibition Road, London, South Kensington SW7 2AZ, United Kingdom
- Rutherford
Appleton Laboratory, Research Complex at
Harwell, Didcot, Oxfordshire OX11 0FA, United Kingdom
| |
Collapse
|
7
|
Preuss F, Chatterjee D, Dederer V, Knapp S, Mathea S. Enabling pseudokinases as potential drug targets. Methods Enzymol 2022; 667:663-683. [PMID: 35525558 DOI: 10.1016/bs.mie.2022.03.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pseudokinases play significant roles in disease development. Similar to active kinases, their cellular functions can be targeted pharmacologically. But notably, instead of inhibiting an enzymatic activity, drug-like molecules act by stabilizing distinct pseudokinase conformations, by interfering with protein interactions, or by inducing proteasomal degradation. Herein, we describe our approach of enabling particular pseudokinases as potential drug targets. The method starts with obtaining recombinant proteins for assay development and for biochemical evaluation. The next step is to probe the pseudoactive site as a binding pocket for small molecules, providing initial insight into binding modes and even candidate chemotypes. Finally, structural features of pseudokinase:inhibitor complexes are explored. Taken together, we provide detailed method descriptions for essential inhibitor development technologies.
Collapse
Affiliation(s)
- Franziska Preuss
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Deep Chatterjee
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Verena Dederer
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany.
| | - Sebastian Mathea
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
8
|
Baltzer S, Bulatov T, Schmied C, Krämer A, Berger BT, Oder A, Walker-Gray R, Kuschke C, Zühlke K, Eichhorst J, Lehmann M, Knapp S, Weston J, von Kries JP, Süssmuth RD, Klussmann E. Aurora Kinase A Is Involved in Controlling the Localization of Aquaporin-2 in Renal Principal Cells. Int J Mol Sci 2022; 23:ijms23020763. [PMID: 35054947 PMCID: PMC8776063 DOI: 10.3390/ijms23020763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 02/01/2023] Open
Abstract
The cAMP-dependent aquaporin-2 (AQP2) redistribution from intracellular vesicles into the plasma membrane of renal collecting duct principal cells induces water reabsorption and fine-tunes body water homeostasis. However, the mechanisms controlling the localization of AQP2 are not understood in detail. Using immortalized mouse medullary collecting duct (MCD4) and primary rat inner medullary collecting duct (IMCD) cells as model systems, we here discovered a key regulatory role of Aurora kinase A (AURKA) in the control of AQP2. The AURKA-selective inhibitor Aurora-A inhibitor I and novel derivatives as well as a structurally different inhibitor, Alisertib, prevented the cAMP-induced redistribution of AQP2. Aurora-A inhibitor I led to a depolymerization of actin stress fibers, which serve as tracks for the translocation of AQP2-bearing vesicles to the plasma membrane. The phosphorylation of cofilin-1 (CFL1) inactivates the actin-depolymerizing function of CFL1. Aurora-A inhibitor I decreased the CFL1 phosphorylation, accounting for the removal of the actin stress fibers and the inhibition of the redistribution of AQP2. Surprisingly, Alisertib caused an increase in actin stress fibers and did not affect CFL1 phosphorylation, indicating that AURKA exerts its control over AQP2 through different mechanisms. An involvement of AURKA and CFL1 in the control of the localization of AQP2 was hitherto unknown.
Collapse
Affiliation(s)
- Sandrine Baltzer
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Timur Bulatov
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Christopher Schmied
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
- DKTK (German Translational Research Network), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| | - Benedict-Tilman Berger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
| | - Andreas Oder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Ryan Walker-Gray
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Christin Kuschke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Kerstin Zühlke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
| | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany; (A.K.); (B.-T.B.); (S.K.)
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
- DKTK (German Translational Research Network), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, 60596 Frankfurt am Main, Germany
| | - John Weston
- JQuest Consulting, Carl-Orff-Weg 25, 65779 Kelkheim, Germany;
| | - Jens Peter von Kries
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (C.S.); (A.O.); (J.E.); (M.L.); (J.P.v.K.)
| | - Roderich D. Süssmuth
- Institute of Chemistry, Technische Universität Berlin, Strasse des 17. Juni 135, 10623 Berlin, Germany; (T.B.); (R.D.S.)
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany; (S.B.); (R.W.-G.); (C.K.); (K.Z.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence: ; Tel.: +49-30-9406-2596
| |
Collapse
|
9
|
Chatterjee D, Preuss F, Dederer V, Knapp S, Mathea S. Structural Aspects of LIMK Regulation and Pharmacology. Cells 2022; 11:cells11010142. [PMID: 35011704 PMCID: PMC8750758 DOI: 10.3390/cells11010142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
Malfunction of the actin cytoskeleton is linked to numerous human diseases including neurological disorders and cancer. LIMK1 (LIM domain kinase 1) and its paralogue LIMK2 are two closely related kinases that control actin cytoskeleton dynamics. Consequently, they are potential therapeutic targets for the treatment of such diseases. In the present review, we describe the LIMK conformational space and its dependence on ligand binding. Furthermore, we explain the unique catalytic mechanism of the kinase, shedding light on substrate recognition and how LIMK activity is regulated. The structural features are evaluated for implications on the drug discovery process. Finally, potential future directions for targeting LIMKs pharmacologically, also beyond just inhibiting the kinase domain, are discussed.
Collapse
Affiliation(s)
- Deep Chatterjee
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str 15, 60438 Frankfurt am Main, Germany; (D.C.); (F.P.); (V.D.); (S.K.)
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str 9, 60438 Frankfurt am Main, Germany
| | - Franziska Preuss
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str 15, 60438 Frankfurt am Main, Germany; (D.C.); (F.P.); (V.D.); (S.K.)
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str 9, 60438 Frankfurt am Main, Germany
| | - Verena Dederer
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str 15, 60438 Frankfurt am Main, Germany; (D.C.); (F.P.); (V.D.); (S.K.)
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str 9, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str 15, 60438 Frankfurt am Main, Germany; (D.C.); (F.P.); (V.D.); (S.K.)
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str 9, 60438 Frankfurt am Main, Germany
| | - Sebastian Mathea
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str 15, 60438 Frankfurt am Main, Germany; (D.C.); (F.P.); (V.D.); (S.K.)
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str 9, 60438 Frankfurt am Main, Germany
- Correspondence:
| |
Collapse
|