1
|
Hu W, Zhang J, Wu Z, Wu Y, Hu Y, Hu X, Cao J. Research progress on paternal mitochondrial inheritance: An overview. Mitochondrion 2025; 82:102019. [PMID: 40024491 DOI: 10.1016/j.mito.2025.102019] [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: 07/05/2024] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Mitochondria are self-replicating organelles with their own DNA. They play a crucial role in biological, cellular and functional processes, such as energy production, metabolism, and signal transduction. Abnormal mitochondrial function can cause various diseases such as diabetes, tumour, Parkinson's disease, hereditary optic neuropathy, and others. Although mitochondrial functions have been extensively and widely explored, studies on mitochondrial inheritance have been limited. Mitochondrial inheritance is traditionally thought to be maternal although small amounts of paternally transmitted mitochondria have been discovered on rare occasions, and the role of paternal mitochondria transmission to offspring has been largely ignored. This review highlights the present knowledge on mitochondrial inheritance, especially the controversy and the difficulties in investigating paternal mitochondrial inheritance. More significantly, we present a comprehensive description of the physiological functions of paternal mitochondria in children and discuss the animal model to explore the mechanism of paternal mitochondrial inheritance. This review may provide a theoretical and experimental basis for improving our understanding of paternal mitochondrial inheritance, and also provide new ideas for treating mitochondrial diseases.
Collapse
Affiliation(s)
- Wen Hu
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jiting Zhang
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Zhaoqi Wu
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yuhui Hu
- Medical College, Jinggangshan University, Ji'an, China
| | - Xiaohui Hu
- Department of Basic Medicine, Gannan Healthcare Vocational College, Ganzhou, China.
| | - Jinguo Cao
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
2
|
Shaw S, Pore SK, Liu D, Kumeria T, Nayak R, Bose S. Combating chemoresistance: Current approaches & nanocarrier mediated targeted delivery. Biochim Biophys Acta Rev Cancer 2025; 1880:189261. [PMID: 39798822 DOI: 10.1016/j.bbcan.2025.189261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Chemoresistance, a significant challenge in effective cancer treatment needs clear elucidation of the underlying molecular mechanism for the development of novel therapeutic strategies. Alterations in transporter pumps, oncogenes, tumour suppressor genes, mitochondrial function, DNA repair processes, autophagy, epithelial-mesenchymal transition (EMT), cancer stemness, epigenetic modifications, and exosome secretion lead to chemoresistance. Despite notable advancements in targeted cancer therapies employing both small molecules and macromolecules success rates remain suboptimal due to adverse effects like drug efflux, target mutation, increased mortality of normal cells, defective apoptosis, etc. This review proposes an advanced nanotechnological technique precisely targeting molecular determinants of chemoresistance which holds promise for enhancing cancer treatment efficacy. Further, the review explores various cancer hallmarks and pathways implicated in chemoresistance, current therapeutic modalities, and their limitations. It advocates the combination of nanoparticle-conjugated conventional drugs and natural compounds to specifically target molecular pathways that can potentially reverse or minimize chemoresistance incidences in cancer patients.
Collapse
Affiliation(s)
- Siuli Shaw
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Subrata Kumar Pore
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Dutong Liu
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University, Noida, Uttar Pradesh, India.
| | - Sudeep Bose
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India; Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India.
| |
Collapse
|
3
|
Xu Y, Peng XL, East MP, McCabe IC, Stroman GC, Jenner MR, Morrison AB, Herrera G, Chan PS, Shen EC, Joisa CU, Rashid NU, Iuga AC, Gomez SM, Miller-Phillips L, Boeck S, Heinemann V, Haas M, Ormanns S, Johnson GL, Yeh JJ. Tumor-Intrinsic Kinome Landscape of Pancreatic Cancer Reveals New Therapeutic Approaches. Cancer Discov 2025; 15:346-362. [PMID: 39632628 PMCID: PMC11805639 DOI: 10.1158/2159-8290.cd-23-1480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
SIGNIFICANCE We provide a comprehensive tumor-intrinsic kinome landscape that provides a roadmap for the use of kinase inhibitors in PDAC treatment approaches.
Collapse
Affiliation(s)
- Yi Xu
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Xianlu L. Peng
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Michael P. East
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Ian C. McCabe
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Grace C. Stroman
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Madison R. Jenner
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Ashley B. Morrison
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Gabriela Herrera
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Priscilla S. Chan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Emily C. Shen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Chinmaya U. Joisa
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Naim U. Rashid
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
- Department of Biostatistics, University of North Carolina at Chapel Hill
| | - Alina C. Iuga
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Shawn M. Gomez
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Lisa Miller-Phillips
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, D-81377, Munich, Germany
- Division of Hematology and Oncology, Department of Medicine and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Stefan Boeck
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, D-81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Volker Heinemann
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, D-81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Michael Haas
- Department of Internal Medicine III and Comprehensive Cancer Center, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, D-81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Steffen Ormanns
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Gary L. Johnson
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Jen Jen Yeh
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
- Department of Surgery, University of North Carolina at Chapel Hill
| |
Collapse
|
4
|
Deng R, Zong GF, Wang X, Yue BJ, Cheng P, Tao RZ, Li X, Wei ZH, Lu Y. Promises of natural products as clinical applications for cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189241. [PMID: 39674416 DOI: 10.1016/j.bbcan.2024.189241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Cancer represents a substantial threat to human health and mortality, necessitating the development of novel pharmacological agents with innovative mechanisms of action. Consequently, extensive research has been directed toward discovering new anticancer compounds derived from natural sources, including plants, microbes, and marine organisms. This review offers a comprehensive analysis of natural anticancer agents that are either currently undergoing clinical trials or have been integrated into clinical practice. A comprehensive understanding of the historical origins of natural anticancer agents, alongside traditional targets for tumor treatment and the distinct characteristics of cancer, can significantly facilitate researchers in the discovery and development of innovative anticancer drugs for clinical use. Furthermore, the exploration of microbial and marine sources is currently a prominent area of focus in the clinical application and advancement of new anticancer therapies. Detailed classification and elucidation of the functions and antitumor properties of these natural products are essential. It is imperative to comprehensively summarize and comprehend the natural anticancer drugs that have been and continue to be utilized in clinical settings.
Collapse
Affiliation(s)
- Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Nanjing Integrated Traditional Chinese And Western Medicine Hospital, Nanjing 210018. China
| | - Gang-Fan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Bing-Jie Yue
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Rui-Zhi Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhong-Hong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023. China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
5
|
Sambataro D, Gebbia V, Bonasera A, Quattrocchi AMO, Caputo G, Vinci E, Di Mattia P, Lavalle S, Pecorino B, Scandurra G, Scibilia G, Centonze D, Valerio MR. Brain Metastasis in Endometrial Cancer: A Systematic Review. Cancers (Basel) 2025; 17:402. [PMID: 39941769 PMCID: PMC11816136 DOI: 10.3390/cancers17030402] [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: 12/04/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Brain metastases (BMs) from endometrial cancer (EC) are rare and challenging to treat, with limited standardized guidelines. This systematic review aims to evaluate the incidence, therapeutic strategies, and outcomes associated with brain metastases in EC patients, offering insights for clinical practice and future research. METHODS A comprehensive literature search was conducted using PRISMA guidelines, including PUBMED up to October 2024. Reports reporting individual or aggregate data on EC brain metastases were included. Descriptive and quantitative analyses were performed on incidence, treatment modalities, and survival outcomes. Three reports that used data from the Surveillance, Epidemiology, and End Results and National Cancer Database were used only to assess the incidence of brain metastases from endometrial carcinoma. RESULTS From 911 reports identified, we included 99 reports, identifying 594 cases; these and the case of a patient with brain metastasis from endometrial carcinoma followed at our center were used for analysis of disease characteristics; incidence; and treatment modalities, such as surgery, radiotherapy, chemotherapy, and combinations. Survival outcomes were influenced by treatment type and disease characteristics, with multimodal approaches showing improved outcomes. DISCUSSION This review underscores the rarity of EC brain metastases and highlights the need for tailored, multimodal treatment strategies. Future research should focus on prospective trials and molecular profiling to optimize management.
Collapse
Affiliation(s)
- Daniela Sambataro
- Medical Oncology Unit, Umberto I Hospital, 94100 Enna, Italy; (A.B.); (A.M.O.Q.); (G.C.); (E.V.)
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
| | - Vittorio Gebbia
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
| | - Annalisa Bonasera
- Medical Oncology Unit, Umberto I Hospital, 94100 Enna, Italy; (A.B.); (A.M.O.Q.); (G.C.); (E.V.)
| | | | - Giuseppe Caputo
- Medical Oncology Unit, Umberto I Hospital, 94100 Enna, Italy; (A.B.); (A.M.O.Q.); (G.C.); (E.V.)
| | - Ernesto Vinci
- Medical Oncology Unit, Umberto I Hospital, 94100 Enna, Italy; (A.B.); (A.M.O.Q.); (G.C.); (E.V.)
| | - Paolo Di Mattia
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
- Surgery Unit, Umberto I Hospital, 94100 Enna, Italy;
| | - Salvatore Lavalle
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
- Diagnostic Imaging Department, Umberto I Hospital, 94100 Enna, Italy
| | - Basilio Pecorino
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
- Gynecology and Obstetrics Unit, Umberto I Hospital, 94100 Enna, Italy
| | - Giuseppa Scandurra
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
- Medical Oncology Unit, Cannizzaro Hospital, 95126 Catania, Italy
| | - Giuseppe Scibilia
- Department of Medicine and Surgery, Kore University, 94100 Enna, Italy; (V.G.); (P.D.M.); (S.L.); (B.P.); (G.S.); (G.S.)
- Gynecology Unit, Giovanni Paolo II Hospital, 97100 Ragusa, Italy
| | | | | |
Collapse
|
6
|
Lucet IS, Daly RJ. View from the PEAKs: Insights from structural studies on the PEAK family of pseudokinases. Curr Opin Struct Biol 2024; 89:102932. [PMID: 39321525 DOI: 10.1016/j.sbi.2024.102932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
The PEAK family of pseudokinase scaffolds, comprising PEAK1 (originally termed SgK269), PEAK2 (SgK223, the human orthologue of rat Pragmin) and PEAK3 (C19orf35), have emerged as important regulators and integrators of cellular signaling and also play oncogenic roles in a variety of human cancers. These proteins undergo both homo- and heterotypic association that act to diversify signal output. Recently, structural and functional characterization of PEAK3 and its protein-protein interactions have shed light on PEAK signaling dynamics and the interdependency of PEAK family members, how PEAK dimerization regulates the binding of downstream effectors, and how 14-3-3 binding acts to regulate PEAK3 signal output. These important advances form the basis of this review.
Collapse
Affiliation(s)
- Isabelle S Lucet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Roger J Daly
- Cancer Program, Biomedical Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
7
|
Han C, Fu S, Chen M, Gou Y, Liu D, Zhang C, Huang X, Xiao L, Zhao M, Zhang J, Xiao Q, Peng D, Xue Y. GPSD: a hybrid learning framework for the prediction of phosphatase-specific dephosphorylation sites. Brief Bioinform 2024; 26:bbae694. [PMID: 39749667 PMCID: PMC11695897 DOI: 10.1093/bib/bbae694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Protein phosphorylation is dynamically and reversibly regulated by protein kinases and protein phosphatases, and plays an essential role in orchestrating a wide range of biological processes. Although a number of tools have been developed for predicting kinase-specific phosphorylation sites (p-sites), computational prediction of phosphatase-specific dephosphorylation sites remains to be a great challenge. In this study, we manually curated 4393 experimentally identified site-specific phosphatase-substrate relationships for 3463 dephosphorylation sites occurring on phosphoserine, phosphothreonine, and/or phosphotyrosine residues, from the literature and public databases. Then, we developed a hybrid learning framework, the group-based prediction system for the prediction of phosphatase-specific dephosphorylation sites (GPSD). For model training, we integrated 10 types of sequence features and utilized three types of machine learning methods, including penalized logistic regression, deep neural networks, and transformer neural networks. First, a pretrained model was constructed using 561 416 nonredundant p-sites and then fine-tuned to generate computational models for predicting general dephosphorylation sites. In addition, 103 individual phosphatase-specific predictors were constructed via transfer learning and meta-learning. For site prediction, one or multiple protein sequences in FASTA format could be inputted, and the prediction results will be shown together with additional annotations, such as protein-protein interactions, structural information, and disorder propensity. The online service of GPSD is freely available at https://gpsd.biocuckoo.cn/. We believe that GPSD can serve as a valuable tool for further analysis of dephosphorylation.
Collapse
Affiliation(s)
- Cheng Han
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Shanshan Fu
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Miaomiao Chen
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Yujie Gou
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Dan Liu
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Chi Zhang
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Xinhe Huang
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Leming Xiao
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Miaoying Zhao
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Jiayi Zhang
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Qiang Xiao
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Di Peng
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| | - Yu Xue
- Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Wuhan, Hubei 430074, China
| |
Collapse
|
8
|
Hillis AL, Tamir T, Perry GE, Asara JM, Johnson JL, Yaron TM, Cantley LC, White FM, Toker A. Parallel phosphoproteomics and metabolomics map the global metabolic tyrosine phosphoproteome. Proc Natl Acad Sci U S A 2024; 121:e2413837121. [PMID: 39536083 PMCID: PMC11588116 DOI: 10.1073/pnas.2413837121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Tyrosine phosphorylation of metabolic enzymes is an evolutionarily conserved posttranslational modification that facilitates rapid and reversible modulation of enzyme activity, localization, or function. Despite the high abundance of tyrosine phosphorylation events detected on metabolic enzymes in high-throughput mass spectrometry-based studies, functional characterization of tyrosine phosphorylation sites has been limited to a subset of enzymes. Since tyrosine phosphorylation is dysregulated across human diseases, including cancer, understanding the consequences of metabolic enzyme tyrosine phosphorylation events is critical for informing disease biology and therapeutic interventions. To globally identify metabolic enzyme tyrosine phosphorylation events and simultaneously assign functional significance to these sites, we performed parallel phosphoproteomics and polar metabolomics in nontumorigenic mammary epithelial cells (MCF10A) stimulated with epidermal growth factor (EGF) in the absence or presence of the EGF receptor inhibitor erlotinib. We performed an integrated analysis of the phosphoproteomic and metabolomic datasets to identify tyrosine phosphorylation sites on metabolic enzymes with functional consequences. We identified two previously characterized (pyruvate kinase muscle isozyme, phosphoglycerate mutase 1) and two uncharacterized (glutathione S-transferase Pi 1, glutamate dehydrogenase 1) tyrosine phosphorylation sites on metabolic enzymes with purported functions based on metabolomic analyses. We validated these hits using a doxycycline-inducible CRISPR interference system in MCF10A cells, in which target metabolic enzymes were depleted with simultaneous reexpression of wild-type, phosphomutant, or phosphomimetic isoforms. Together, these data provide a framework for identification, prioritization, and characterization of tyrosine phosphorylation sites on metabolic enzymes with functional significance.
Collapse
Affiliation(s)
- Alissandra L. Hillis
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Tigist Tamir
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Grace E. Perry
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - John M. Asara
- Mass Spectrometry Core, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jared L. Johnson
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Tomer M. Yaron
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Lewis C. Cantley
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Forest M. White
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
9
|
Beyer JN, Serebrenik YV, Toy K, Najar MA, Raniszewski NR, Shalem O, Burslem GM. Intracellular Protein Editing to Enable Incorporation of Non-Canonical Residues into Endogenous Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602493. [PMID: 39026884 PMCID: PMC11257474 DOI: 10.1101/2024.07.08.602493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The ability to study proteins in a cellular context is crucial to our understanding of biology. Here, we report a new technology for "intracellular protein editing", drawing from intein- mediated protein splicing, genetic code expansion, and endogenous protein tagging. This protein editing approach enables us to rapidly and site specifically install residues and chemical handles into a protein of interest. We demonstrate the power of this protein editing platform to edit cellular proteins, inserting epitope peptides, protein-specific sequences, and non-canonical amino acids (ncAAs). Importantly, we employ an endogenous tagging approach to apply our protein editing technology to endogenous proteins with minimal perturbation. We anticipate that the protein editing technology presented here will be applied to a diverse set of problems, enabling novel experiments in live mammalian cells and therefore provide unique biological insights.
Collapse
|
10
|
Royet C, Diot S, Onofre M, Lecki L, Pastore M, Reynes C, Lorcy F, Lacheretzszablewski V, Serre I, Morris MC. Multiplexed Profiling of CDK Kinase Activities in Tumor Biopsies with Fluorescent Peptide Biosensors. ACS Sens 2024; 9:2964-2978. [PMID: 38863434 DOI: 10.1021/acssensors.4c00139] [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] [Indexed: 06/13/2024]
Abstract
Detection of disease biomarkers constitutes a major challenge for the development of personalized and predictive diagnostics as well as companion assays. Protein kinases (PKs) involved in the coordination of cell cycle progression and proliferation that are hyperactivated in human cancers constitute attractive pharmacological targets and relevant biomarkers. Although it is relatively straightforward to assess the relative abundance of PKs in a biological sample, there is not always a direct correlation with enzymatic activity, which is regulated by several posttranslational mechanisms. Studies of relative abundance therefore convey limited information, and the lack of selective, sensitive, and standardized tools together with the inherent complexity of biological samples makes it difficult to quantify PK activities in physio-pathological tissues. To address this challenge, we have developed a toolbox of fluorescent biosensors that report on CDK activities in a sensitive, selective, dose-dependent, and quantitative fashion, which we have implemented to profile CDK activity signatures in cancer cell lines and biopsies from human tumors. In this study, we report on a standardized and calibrated biosensing approach to quantify CDK1,2,4, and 6 activities simultaneously through a combination of four different biosensors in a panel of 40 lung adenocarcinoma and 40 follicular lymphoma samples. CDK activity profiling highlighted two major patterns which were further correlated with age, sex of patients, tumor size, grade, and genetic and immunohistochemical features of the biopsies. Multiplex CDKACT biosensing technology provides new and complementary information relative to current genetic and immunohistochemical characterization of tumor biopsies, which will be useful for diagnostic purposes, potentially guiding therapeutic decision. These fluorescent peptide biosensors offer promise for personalized diagnostics based on kinase activity profiling.
Collapse
Affiliation(s)
- Chloé Royet
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Montpellier University, 1919 Route de Mende, 34293 Montpellier, France
| | - Sébastien Diot
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Montpellier University, 1919 Route de Mende, 34293 Montpellier, France
| | - Mélanie Onofre
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Montpellier University, 1919 Route de Mende, 34293 Montpellier, France
| | - Lennard Lecki
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Montpellier University, 1919 Route de Mende, 34293 Montpellier, France
| | - Manuela Pastore
- StatABio Facility─Biocampus, UAR 3426 CNRS─US 09 INSERM, Montpellier University, 141 rue de la Cardonille, 34094 Montpellier Cedex 05, France
| | - Christelle Reynes
- StatABio Facility─Biocampus, UAR 3426 CNRS─US 09 INSERM, Montpellier University, 141 rue de la Cardonille, 34094 Montpellier Cedex 05, France
| | - Frederique Lorcy
- University Hospital Centre Montpellier, 80 Av. Augustin Fliche, 34295 Montpellier, France
| | | | - Isabelle Serre
- University Hospital Centre Montpellier, 80 Av. Augustin Fliche, 34295 Montpellier, France
| | - May C Morris
- Institut des Biomolécules Max Mousseron, CNRS, UMR 5247, Montpellier University, 1919 Route de Mende, 34293 Montpellier, France
| |
Collapse
|
11
|
Dalle S. Targeting Protein Kinases to Protect Beta-Cell Function and Survival in Diabetes. Int J Mol Sci 2024; 25:6425. [PMID: 38928130 PMCID: PMC11203834 DOI: 10.3390/ijms25126425] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of diabetes is increasing worldwide. Massive death of pancreatic beta-cells causes type 1 diabetes. Progressive loss of beta-cell function and mass characterizes type 2 diabetes. To date, none of the available antidiabetic drugs promotes the maintenance of a functional mass of endogenous beta-cells, revealing an unmet medical need. Dysfunction and apoptotic death of beta-cells occur, in particular, through the activation of intracellular protein kinases. In recent years, protein kinases have become highly studied targets of the pharmaceutical industry for drug development. A number of drugs that inhibit protein kinases have been approved for the treatment of cancers. The question of whether safe drugs that inhibit protein kinase activity can be developed and used to protect the function and survival of beta-cells in diabetes is still unresolved. This review presents arguments suggesting that several protein kinases in beta-cells may represent targets of interest for the development of drugs to treat diabetes.
Collapse
Affiliation(s)
- Stéphane Dalle
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 34094 Montpellier, France
| |
Collapse
|
12
|
Bazant J, Weiss A, Baldauf J, Schermuly RT, Hain T, Lucas R, Mraheil MA. Pneumococcal hydrogen peroxide regulates host cell kinase activity. Front Immunol 2024; 15:1414195. [PMID: 38903521 PMCID: PMC11188345 DOI: 10.3389/fimmu.2024.1414195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction Protein kinases are indispensable reversible molecular switches that adapt and control protein functions during cellular processes requiring rapid responses to internal and external events. Bacterial infections can affect kinase-mediated phosphorylation events, with consequences for both innate and adaptive immunity, through regulation of antigen presentation, pathogen recognition, cell invasiveness and phagocytosis. Streptococcus pneumoniae (Spn), a human respiratory tract pathogen and a major cause of community-acquired pneumoniae, affects phosphorylation-based signalling of several kinases, but the pneumococcal mediator(s) involved in this process remain elusive. In this study, we investigated the influence of pneumococcal H2O2 on the protein kinase activity of the human lung epithelial H441 cell line, a generally accepted model of alveolar epithelial cells. Methods We performed kinome analysis using PamGene microarray chips and protein analysis in Western blotting in H441 lung cells infected with Spn wild type (SpnWT) or with SpnΔlctOΔspxB -a deletion mutant strongly attenuated in H2O2 production- to assess the impact of pneumococcal hydrogen peroxide (H2O2) on global protein kinase activity profiles. Results Our kinome analysis provides direct evidence that kinase activity profiles in infected H441 cells significantly vary according to the levels of pneumococcal H2O2. A large number of kinases in H441 cells infected with SpnWT are significantly downregulated, whereas this no longer occurs in cells infected with the mutant SpnΔlctOΔspxB strain, which lacks H2O2. In particular, we describe for the first time H2O2-mediated downregulation of Protein kinase B (Akt1) and activation of lymphocyte-specific tyrosine protein kinase (Lck) via H2O2-mediated phosphorylation.
Collapse
Affiliation(s)
- Jasmin Bazant
- Institute of Medical Microbiology, German Centre for Infection Giessen-Marburg-Langen Site, Justus-Liebig University Giessen, Giessen, Germany
| | - Astrid Weiss
- Department of Internal Medicine, Cardio–Pulmonary Institute (CPI), Member of German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Julia Baldauf
- Department of Internal Medicine, Cardio–Pulmonary Institute (CPI), Member of German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Cardio–Pulmonary Institute (CPI), Member of German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Torsten Hain
- Institute of Medical Microbiology, German Centre for Infection Giessen-Marburg-Langen Site, Justus-Liebig University Giessen, Giessen, Germany
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Division of Pulmonary, Sleep and Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Mobarak Abu Mraheil
- Institute of Medical Microbiology, German Centre for Infection Giessen-Marburg-Langen Site, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
13
|
Mariella E, Grasso G, Miotto M, Buzo K, Reilly NM, Andrei P, Vitiello PP, Crisafulli G, Arena S, Rospo G, Corti G, Lorenzato A, Cancelliere C, Barault L, Gionfriddo G, Linnebacher M, Russo M, Di Nicolantonio F, Bardelli A. Transcriptome-wide gene expression outlier analysis pinpoints therapeutic vulnerabilities in colorectal cancer. Mol Oncol 2024; 18:1460-1485. [PMID: 38468448 PMCID: PMC11161737 DOI: 10.1002/1878-0261.13622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
Multiple strategies are continuously being explored to expand the drug target repertoire in solid tumors. We devised a novel computational workflow for transcriptome-wide gene expression outlier analysis that allows the systematic identification of both overexpression and underexpression events in cancer cells. Here, it was applied to expression values obtained through RNA sequencing in 226 colorectal cancer (CRC) cell lines that were also characterized by whole-exome sequencing and microarray-based DNA methylation profiling. We found cell models displaying an abnormally high or low expression level for 3533 and 965 genes, respectively. Gene expression abnormalities that have been previously associated with clinically relevant features of CRC cell lines were confirmed. Moreover, by integrating multi-omics data, we identified both genetic and epigenetic alternations underlying outlier expression values. Importantly, our atlas of CRC gene expression outliers can guide the discovery of novel drug targets and biomarkers. As a proof of concept, we found that CRC cell lines lacking expression of the MTAP gene are sensitive to treatment with a PRMT5-MTA inhibitor (MRTX1719). Finally, other tumor types may also benefit from this approach.
Collapse
Affiliation(s)
- Elisa Mariella
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
- IFOM ETS, The AIRC Institute of Molecular OncologyMilanItaly
| | - Gaia Grasso
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
- IFOM ETS, The AIRC Institute of Molecular OncologyMilanItaly
| | - Martina Miotto
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
- IFOM ETS, The AIRC Institute of Molecular OncologyMilanItaly
| | - Kristi Buzo
- Department of OncologyUniversity of TorinoCandiolo (TO)Italy
- Candiolo Cancer InstituteFPO‐IRCCSCandiolo (TO)Italy
| | | | - Pietro Andrei
- Department of OncologyUniversity of TorinoCandiolo (TO)Italy
| | - Pietro Paolo Vitiello
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
- IFOM ETS, The AIRC Institute of Molecular OncologyMilanItaly
| | | | - Sabrina Arena
- Department of OncologyUniversity of TorinoCandiolo (TO)Italy
- Candiolo Cancer InstituteFPO‐IRCCSCandiolo (TO)Italy
| | - Giuseppe Rospo
- Department of OncologyUniversity of TorinoCandiolo (TO)Italy
- Present address:
Boehringer Ingelheim RCV GmbH & Co KGViennaAustria
| | - Giorgio Corti
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Annalisa Lorenzato
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
| | | | - Ludovic Barault
- Department of OncologyUniversity of TorinoCandiolo (TO)Italy
| | | | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and ImmunotherapyUniversity of RostockGermany
| | - Mariangela Russo
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
- IFOM ETS, The AIRC Institute of Molecular OncologyMilanItaly
| | - Federica Di Nicolantonio
- Department of OncologyUniversity of TorinoCandiolo (TO)Italy
- Candiolo Cancer InstituteFPO‐IRCCSCandiolo (TO)Italy
| | - Alberto Bardelli
- Department of Oncology, Molecular Biotechnology CenterUniversity of TorinoItaly
- IFOM ETS, The AIRC Institute of Molecular OncologyMilanItaly
| |
Collapse
|
14
|
Yao CY, Lin CC, Wang YH, Kao CJ, Tsai CH, Hou HA, Tien HF, Hsu CL, Chou WC. Kinome expression profiling improves risk stratification and therapeutic targeting in myelodysplastic syndromes. Blood Adv 2024; 8:2442-2454. [PMID: 38527292 PMCID: PMC11112608 DOI: 10.1182/bloodadvances.2023011512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 03/27/2024] Open
Abstract
ABSTRACT The human kinome, which comprises >500 kinases, plays a critical role in regulating numerous essential cellular functions. Although the dysregulation of kinases has been observed in various human cancers, the characterization and clinical implications of kinase expressions in myelodysplastic syndromes (MDS) have not been systematically investigated. In this study, we evaluated the kinome expression profiles of 341 adult patients with primary MDS and identified 7 kinases (PTK7, KIT, MAST4, NTRK1, PAK6, CAMK1D, and PRKCZ) whose expression levels were highly predictive of compromised patient survival. We then constructed the kinase stratification score (KISS) by combining the weighted expressions of the 7 kinases and validated its prognostic significance in 2 external MDS cohorts. A higher KISS was associated with older age, higher peripheral blood and marrow blast percentages, higher Revised International Prognostic Scoring System (IPSS-R) risks, complex karyotype, and mutations in several adverse-risk genes in MDS, such as ASXL1, EZH2, NPM1, RUNX1, STAG2, and TP53. Multivariate analysis confirmed that a higher KISS was an independent unfavorable risk factor in MDS. Mechanistically, the KISS-high patients were enriched for gene sets associated with hematopoietic and leukemic stem cell signatures. By investigating the Genomics of Drug Sensitivity in Cancer database, we identified axitinib and taselisib as candidate compounds that could potentially target the KISS-high myeloblasts. Altogether, our findings suggest that KISS holds the potential to improve the current prognostic scheme of MDS and inform novel therapeutic opportunities.
Collapse
Affiliation(s)
- Chi-Yuan Yao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chin Lin
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Hung Wang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Chein-Jun Kao
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Hong Tsai
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Prus G, Satpathy S, Weinert BT, Narita T, Choudhary C. Global, site-resolved analysis of ubiquitylation occupancy and turnover rate reveals systems properties. Cell 2024; 187:2875-2892.e21. [PMID: 38626770 PMCID: PMC11136510 DOI: 10.1016/j.cell.2024.03.024] [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: 12/19/2023] [Accepted: 03/19/2024] [Indexed: 04/18/2024]
Abstract
Ubiquitylation regulates most proteins and biological processes in a eukaryotic cell. However, the site-specific occupancy (stoichiometry) and turnover rate of ubiquitylation have not been quantified. Here we present an integrated picture of the global ubiquitylation site occupancy and half-life. Ubiquitylation site occupancy spans over four orders of magnitude, but the median ubiquitylation site occupancy is three orders of magnitude lower than that of phosphorylation. The occupancy, turnover rate, and regulation of sites by proteasome inhibitors are strongly interrelated, and these attributes distinguish sites involved in proteasomal degradation and cellular signaling. Sites in structured protein regions exhibit longer half-lives and stronger upregulation by proteasome inhibitors than sites in unstructured regions. Importantly, we discovered a surveillance mechanism that rapidly and site-indiscriminately deubiquitylates all ubiquitin-specific E1 and E2 enzymes, protecting them against accumulation of bystander ubiquitylation. The work provides a systems-scale, quantitative view of ubiquitylation properties and reveals general principles of ubiquitylation-dependent governance.
Collapse
Affiliation(s)
- Gabriela Prus
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Shankha Satpathy
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Brian T Weinert
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Takeo Narita
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Chunaram Choudhary
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
16
|
Munteanu RA, Tigu AB, Feder R, Tatar AS, Gulei D, Tomuleasa C, Boca S. In vivo imaging system (IVIS) therapeutic assessment of tyrosine kinase inhibitor-loaded gold nanocarriers for acute myeloid leukemia: a pilot study. Front Pharmacol 2024; 15:1382399. [PMID: 38799169 PMCID: PMC11116693 DOI: 10.3389/fphar.2024.1382399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Acute myeloid leukemia (AML) is a malignancy in the myeloid lineage that is characterized by symptoms like fatigue, bleeding, infections, or anemia, and it can be fatal if untreated. In AML, mutations in tyrosine kinases (TKs) lead to enhanced tumor cell survival. The most frequent mutations in TKs are reported in Fms-like tyrosine kinase 3 (FLT3), Janus kinase 2 (JAK2), and KIT (tyrosine-protein kinase KIT), making these TKs potential targets for TK inhibitor (TKI) therapies in AML. With 30% of the mutations in TKs, mutated FLT3 is associated with poor overall survival and an increased chance of resistance to therapy. FLT3 inhibitors are used in FLT3-mutant AML, and the combination with hypomethylating agents displayed promising results. Midostaurin (MDS) is the first targeted therapy in FLT3-mutant AML, and its combination with chemotherapy showed good results. However, chemotherapies induce several side effects, and an alternative to chemotherapy might be the use of nanoparticles for better drug delivery, improved bioavailability, reduced drug resistance and induced toxicity. The herein study presents MDS-loaded gold nanoparticles and compares its efficacy with MDS alone, on both in vitro and in vivo models, using the FLT3-ITD-mutated AML cell line MV-4-11 Luc2 transfected to express luciferin. Our preclinical study suggests that MDS-loaded nanoparticles have a better tumor inhibitory effect than free drugs on in vivo models by controlling tumor growth in the first half of the treatment, while in the second part of the therapy, the tumor size was comparable to the cohort that was treatment-free.
Collapse
Affiliation(s)
- Raluca-Andrada Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Richard Feder
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andra-Sorina Tatar
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
- Molecular and Biomolecular Physics Department, National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, Cluj-Napoca, Romania
| | - Sanda Boca
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
- Molecular and Biomolecular Physics Department, National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| |
Collapse
|
17
|
Reveguk I, Simonson T. Classifying protein kinase conformations with machine learning. Protein Sci 2024; 33:e4918. [PMID: 38501429 PMCID: PMC10962494 DOI: 10.1002/pro.4918] [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: 07/26/2023] [Revised: 01/02/2024] [Accepted: 01/22/2024] [Indexed: 03/20/2024]
Abstract
Protein kinases are key actors of signaling networks and important drug targets. They cycle between active and inactive conformations, distinguished by a few elements within the catalytic domain. One is the activation loop, whose conserved DFG motif can occupy DFG-in, DFG-out, and some rarer conformations. Annotation and classification of the structural kinome are important, as different conformations can be targeted by different inhibitors and activators. Valuable resources exist; however, large-scale applications will benefit from increased automation and interpretability of structural annotation. Interpretable machine learning models are described for this purpose, based on ensembles of decision trees. To train them, a set of catalytic domain sequences and structures was collected, somewhat larger and more diverse than existing resources. The structures were clustered based on the DFG conformation and manually annotated. They were then used as training input. Two main models were constructed, which distinguished active/inactive and in/out/other DFG conformations. They considered initially 1692 structural variables, spanning the whole catalytic domain, then identified ("learned") a small subset that sufficed for accurate classification. The first model correctly labeled all but 3 of 3289 structures as active or inactive, while the second assigned the correct DFG label to all but 17 of 8826 structures. The most potent classifying variables were all related to well-known structural elements in or near the activation loop and their ranking gives insights into the conformational preferences. The models were used to automatically annotate 3850 kinase structures predicted recently with the Alphafold2 tool, showing that Alphafold2 reproduced the active/inactive but not the DFG-in proportions seen in the Protein Data Bank. We expect the models will be useful for understanding and engineering kinases.
Collapse
Affiliation(s)
- Ivan Reveguk
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654)Ecole PolytechniquePalaiseauFrance
| | - Thomas Simonson
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654)Ecole PolytechniquePalaiseauFrance
| |
Collapse
|
18
|
Stephenson EH, Higgins JMG. Pharmacological approaches to understanding protein kinase signaling networks. Front Pharmacol 2023; 14:1310135. [PMID: 38164473 PMCID: PMC10757940 DOI: 10.3389/fphar.2023.1310135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Protein kinases play vital roles in controlling cell behavior, and an array of kinase inhibitors are used successfully for treatment of disease. Typical drug development pipelines involve biological studies to validate a protein kinase target, followed by the identification of small molecules that effectively inhibit this target in cells, animal models, and patients. However, it is clear that protein kinases operate within complex signaling networks. These networks increase the resilience of signaling pathways, which can render cells relatively insensitive to inhibition of a single kinase, and provide the potential for pathway rewiring, which can result in resistance to therapy. It is therefore vital to understand the properties of kinase signaling networks in health and disease so that we can design effective multi-targeted drugs or combinations of drugs. Here, we outline how pharmacological and chemo-genetic approaches can contribute to such knowledge, despite the known low selectivity of many kinase inhibitors. We discuss how detailed profiling of target engagement by kinase inhibitors can underpin these studies; how chemical probes can be used to uncover kinase-substrate relationships, and how these tools can be used to gain insight into the configuration and function of kinase signaling networks.
Collapse
Affiliation(s)
| | - Jonathan M. G. Higgins
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle uponTyne, United Kingdom
| |
Collapse
|
19
|
Liu C, Kutchukian P, Nguyen ND, AlQuraishi M, Sorger PK. A Hybrid Structure-Based Machine Learning Approach for Predicting Kinase Inhibition by Small Molecules. J Chem Inf Model 2023; 63:5457-5472. [PMID: 37595065 PMCID: PMC10498990 DOI: 10.1021/acs.jcim.3c00347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 08/20/2023]
Abstract
Kinases have been the focus of drug discovery programs for three decades leading to over 70 therapeutic kinase inhibitors and biophysical affinity measurements for over 130,000 kinase-compound pairs. Nonetheless, the precise target spectrum for many kinases remains only partly understood. In this study, we describe a computational approach to unlocking qualitative and quantitative kinome-wide binding measurements for structure-based machine learning. Our study has three components: (i) a Kinase Inhibitor Complex (KinCo) data set comprising in silico predicted kinase structures paired with experimental binding constants, (ii) a machine learning loss function that integrates qualitative and quantitative data for model training, and (iii) a structure-based machine learning model trained on KinCo. We show that our approach outperforms methods trained on crystal structures alone in predicting binary and quantitative kinase-compound interaction affinities; relative to structure-free methods, our approach also captures known kinase biochemistry and more successfully generalizes to distant kinase sequences and compound scaffolds.
Collapse
Affiliation(s)
- Changchang Liu
- Laboratory
of Systems Pharmacology, Department of Systems Biology, Harvard Program
in Therapeutic Science, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Peter Kutchukian
- Novartis
Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Nhan D. Nguyen
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United
States
| | - Mohammed AlQuraishi
- Department
of Systems Biology, Columbia University, New York, New York 10032, United States
| | - Peter K. Sorger
- Laboratory
of Systems Pharmacology, Department of Systems Biology, Harvard Program
in Therapeutic Science, Harvard Medical
School, Boston, Massachusetts 02115, United States
| |
Collapse
|
20
|
Motomura K, Kuwano A, Tanaka K, Koga Y, Masumoto A, Yada M. Potential Predictive Biomarkers of Systemic Drug Therapy for Hepatocellular Carcinoma: Anticipated Usefulness in Clinical Practice. Cancers (Basel) 2023; 15:4345. [PMID: 37686621 PMCID: PMC10486942 DOI: 10.3390/cancers15174345] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
In the systemic drug treatment of hepatocellular carcinoma, only the tyrosine kinase inhibitor (TKI) sorafenib was available for a period. This was followed by the development of regorafenib as a second-line treatment after sorafenib, and then lenvatinib, a new TKI, proved non-inferiority to sorafenib and became available as a first-line treatment. Subsequently, cabozantinib, another TKI, was introduced as a second-line treatment, along with ramucirumab, the only drug proven to be predictive of therapeutic efficacy when AFP levels are >400 ng/mL. It is an anti-VEGF receptor antibody. More recently, immune checkpoint inhibitors have become the mainstay of systemic therapy and can now be used as a first-line standard treatment for HCC. However, the objective response rate for these drugs is currently only 30% to 40%, and there is a high incidence of side effects. Additionally, there are no practical biomarkers to predict their therapeutic effects. Therefore, this review provides an overview of extensive research conducted on potential HCC biomarkers from blood, tissue, or imaging information that can be used in practice to predict the therapeutic efficacy of systemic therapy before its initiation.
Collapse
Affiliation(s)
- Kenta Motomura
- Department of Hepatology, Iizuka Hospital, 3-83 Yoshio-machi, Iizuka, Fukuoka 820-8505, Japan; (A.K.); (K.T.); (Y.K.); (A.M.); (M.Y.)
| | | | | | | | | | | |
Collapse
|
21
|
Messana I, Manconi B, Cabras T, Boroumand M, Sanna MT, Iavarone F, Olianas A, Desiderio C, Rossetti DV, Vincenzoni F, Contini C, Guadalupi G, Fiorita A, Faa G, Castagnola M. The Post-Translational Modifications of Human Salivary Peptides and Proteins Evidenced by Top-Down Platforms. Int J Mol Sci 2023; 24:12776. [PMID: 37628956 PMCID: PMC10454625 DOI: 10.3390/ijms241612776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
In this review, we extensively describe the main post-translational modifications that give rise to the multiple proteoforms characterized to date in the human salivary proteome and their potential role. Most of the data reported were obtained by our group in over twenty-five years of research carried out on human saliva mainly by applying a top-down strategy. In the beginning, we describe the products generated by proteolytic cleavages, which can occur before and after secretion. In this section, the most relevant families of salivary proteins are also described. Next, we report the current information concerning the human salivary phospho-proteome and the limited news available on sulfo-proteomes. Three sections are dedicated to the description of glycation and enzymatic glycosylation. Citrullination and N- and C-terminal post-translational modifications (PTMs) and miscellaneous other modifications are described in the last two sections. Results highlighting the variation in the level of some proteoforms in local or systemic pathologies are also reviewed throughout the sections of the manuscript to underline the impact and relevance of this information for the development of new diagnostic biomarkers useful in clinical practice.
Collapse
Affiliation(s)
- Irene Messana
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Tiziana Cabras
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | | | - Maria Teresa Sanna
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.I.); (F.V.)
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
| | - Alessandra Olianas
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Diana Valeria Rossetti
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Federica Vincenzoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.I.); (F.V.)
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
| | - Cristina Contini
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Giulia Guadalupi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Antonella Fiorita
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
- Dipartimento di Scienze dell’Invecchiamento, Neurologiche, Ortopediche e della Testa e del Collo, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gavino Faa
- Unit of Pathology, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Massimo Castagnola
- Proteomics Laboratory, European Center for Brain Research, (IRCCS) Santa Lucia Foundation, 00168 Rome, Italy;
| |
Collapse
|
22
|
Akula S, Mullaguri SC, Melton NM, Katta A, Naga VSGR, Kandula S, Pedada RK, Subramanian J, Kancha RK. Large-scale pathogenicity prediction analysis of cancer-associated kinase mutations reveals variability in sensitivity and specificity of computational methods. Cancer Med 2023; 12:17468-17474. [PMID: 37409618 PMCID: PMC10501281 DOI: 10.1002/cam4.6324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/26/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Mutations in kinases are the most frequent genetic alterations in cancer; however, experimental evidence establishing their cancerous nature is available only for a small fraction of these mutants. AIMS Predicition analysis of kinome mutations is the primary aim of this study. Further objective is to compare the performance of various softwares in pathogenicity prediction of kinase mutations. MATERIALS AND METHODS We employed a set of computational tools to predict the pathogenicity of over forty-two thousand mutations and deposited the kinase-wise data in Mendeley database (Estimated Pathogenicity of Kinase Mutants [EPKiMu]). RESULTS Mutations are more likely to be drivers when being present in the kinase domain (vs. non-kinase domain) and belonging to hotspot residues (vs. non-hotspot residues). We identified that, while predictive tools have low specificity in general, PolyPhen-2 had the best accuracy. Further efforts to combine all four tools by consensus, voting, or other simple methods did not significantly improve accuracy. DISCUSSION The study provides a large dataset of kinase mutations along with their predicted pathogenicity that can be used as a training set for future studies. Furthermore, a comparative sensitivity and selectivity of commonly used computational tools is presented. CONCLUSION Primary-structure-based in silico tools identified more cancerous/deleterious mutations in the kinase domains and at the hot spot residues while having higher sensitivity than specificity in detecting deleterious mutations.
Collapse
Affiliation(s)
- Sravani Akula
- Molecular Medicine and Therapeutics Laboratory, CPMBOsmania UniversityHyderabadIndia
| | | | - Niklas Max Melton
- Thoracic Oncology, Inova Schar Cancer InstituteFairfaxVirginiaUSA
- Applied Computational Intelligence LabMissouri University of Science and TechnologyRollaMissouriUSA
| | - Archana Katta
- Molecular Medicine and Therapeutics Laboratory, CPMBOsmania UniversityHyderabadIndia
| | | | - Shyamson Kandula
- Molecular Medicine and Therapeutics Laboratory, CPMBOsmania UniversityHyderabadIndia
| | - Raj Kumar Pedada
- Molecular Medicine and Therapeutics Laboratory, CPMBOsmania UniversityHyderabadIndia
| | | | - Rama Krishna Kancha
- Molecular Medicine and Therapeutics Laboratory, CPMBOsmania UniversityHyderabadIndia
| |
Collapse
|
23
|
Chen X, Sun Z, Zhou S, Jiang W, Li J, Song G, Zhu X. SH3 domain-binding kinase 1 promotes proliferation and inhibits apoptosis of cervical cancer via activating the Wnt/β-catenin and Raf/ERK1/2 signaling pathways. Mol Carcinog 2023; 62:1147-1162. [PMID: 37132991 DOI: 10.1002/mc.23552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
SH3 domain-binding kinase 1 (SBK1), is a member of the serine/threonine protein kinases family, and was confirmed to be upregulated in cervical cancer in our previous study. Nonetheless, the role of SBK1 in regulating cancer occurrence and development is unclear. In this study, the stable SBK1-knockdown and -overexpressed cell models were constructed by plasmid transfection technology. Cell viability and growth were assessed through CCK-8, colony formation, and BrdU methods. Cell cycle and apoptosis were analyzed by flow cytometry. The JC-1 staining assay was used to explore mitochondrial membrane potential. The scratch and Transwell assays were used to evaluate the cell metastatic ability. The nude mice models were utilized to explore the SBK1 expression affecting tumor growth in vivo. Our research indicated a high expression of SBK1 both in tissues and cells of cervical cancer. The proliferative, migratory, as well as invasive capacities of cervical cancer cells, were suppressed, and apoptosis was enhanced after SBK1 silence, whereas SBK1 upregulation led to opposite results. In addition, Wnt/β-catenin and Raf/ERK1/2 pathways were activated by SBK1 upregulation. Furthermore, downregulation of c-Raf or β-catenin, reversed the proliferation promotion and apoptosis inhibition effects in SBK1-overexpressed cells. The same results were observed with the use of the specific Raf inhibitor. SBK1 overexpression also contributed to tumor growth in vivo. Overall, SBK1 played a vital role in cervical tumorigenesis via activating the Wnt/β-catenin and Raf/ERK1/2 pathways.
Collapse
Affiliation(s)
- Xin Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengwei Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengjie Zhou
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenxiao Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jieyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gendi Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
24
|
Morita M, Nishida N, Aoki T, Chishina H, Takita M, Ida H, Hagiwara S, Minami Y, Ueshima K, Kudo M. Role of β-Catenin Activation in the Tumor Immune Microenvironment and Immunotherapy of Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:cancers15082311. [PMID: 37190239 DOI: 10.3390/cancers15082311] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Recently, the therapeutic combination of atezolizumab and bevacizumab was widely used to treat advanced hepatocellular carcinoma (HCC). According to recent clinical trials, immune checkpoint inhibitors (ICIs) and molecular target agents are expected to be key therapeutic strategies in the future. Nonetheless, the mechanisms underlying molecular immune responses and immune evasion remain unclear. The tumor immune microenvironment plays a vital role in HCC progression. The infiltration of CD8-positive cells into tumors and the expression of immune checkpoint molecules are key factors in this immune microenvironment. Specifically, Wnt/β catenin pathway activation causes "immune exclusion", associated with poor infiltration of CD8-positive cells. Some clinical studies suggested an association between ICI resistance and β-catenin activation in HCC. Additionally, several subclassifications of the tumor immune microenvironment were proposed. The HCC immune microenvironment can be broadly divided into inflamed class and non-inflamed class, with several subclasses. β-catenin mutations are important factors in immune subclasses; this may be useful when considering therapeutic strategies as β-catenin activation may serve as a biomarker for ICI. Various types of β-catenin modulators were developed. Several kinases may also be involved in the β-catenin pathway. Therefore, combinations of β-catenin modulators, kinase inhibitors, and ICIs may exert synergistic effects.
Collapse
Affiliation(s)
- Masahiro Morita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Tomoko Aoki
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Hirokazu Chishina
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Masahiro Takita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Hiroshi Ida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Satoru Hagiwara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Yasunori Minami
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Kazuomi Ueshima
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| |
Collapse
|
25
|
Yang G, Zuo C, Lin Y, Zhou X, Wen P, Zhang C, Xiao H, Jiang M, Fujita M, Gao XD, Fu F. Comprehensive proteome, phosphoproteome and kinome characterization of luminal A breast cancer. Front Oncol 2023; 13:1127446. [PMID: 37064116 PMCID: PMC10102592 DOI: 10.3389/fonc.2023.1127446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/08/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundBreast cancer is one of the most frequently occurring malignant cancers worldwide. Invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) are the two most common histological subtypes of breast cancer. In this study, we aimed to deeply explore molecular characteristics and the relationship between IDC and ILC subtypes in luminal A subgroup of breast cancer using comprehensive proteomics and phosphoproteomics analysis.MethodsCancer tissues and noncancerous adjacent tissues (NATs) with the luminal A subtype (ER- and PR-positive, HER2-negative) were obtained from paired IDC and ILC patients respectively. Label-free quantitative proteomics and phosphoproteomics methods were used to detect differential proteins and the phosphorylation status between 10 paired breast cancer and NATs. Then, the difference in protein expression and its phosphorylation between IDC and ILC subtypes were explored. Meanwhile, the activation of kinases and their substrates was also revealed by Kinase-Substrate Enrichment Analysis (KSEA).ResultsIn the luminal A breast cancer, a total of 5,044 high-confidence proteins and 3,808 phosphoproteins were identified from 10 paired tissues. The protein phosphorylation level in ILC tissues was higher than that in IDC tissues. Histone H1.10 was significantly increased in IDC but decreased in ILC, Conversely, complement C4-B and Crk-like protein were significantly decreased in IDC but increased in ILC. Moreover, the increased protein expression of Septin-2, Septin-9, Heterogeneous nuclear ribonucleoprotein A1 and Kinectin but reduce of their phosphorylation could clearly distinguish IDC from ILC. In addition, IDC was primarily related to energy metabolism and MAPK pathway, while ILC was more closely involved in the AMPK and p53/p21 pathways. Furthermore, the kinomes in IDC were primarily significantly activated in the CMGC groups.ConclusionsOur research provides insights into the molecular characterization of IDC and ILC and contributes to discovering novel targets for further drug development and targeted treatment.
Collapse
Affiliation(s)
- Ganglong Yang
- The Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Chenyang Zuo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Yuxiang Lin
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoman Zhou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Piaopiao Wen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Chairui Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Han Xiao
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Meichen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Morihisa Fujita
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Xiao-Dong Gao
- The Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- *Correspondence: Xiao-Dong Gao, ; Fangmeng Fu,
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian, China
- *Correspondence: Xiao-Dong Gao, ; Fangmeng Fu,
| |
Collapse
|
26
|
Viganò M, La Milia M, Grassini MV, Pugliese N, De Giorgio M, Fagiuoli S. Hepatotoxicity of Small Molecule Protein Kinase Inhibitors for Cancer. Cancers (Basel) 2023; 15:cancers15061766. [PMID: 36980652 PMCID: PMC10046041 DOI: 10.3390/cancers15061766] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Small molecule protein kinase inhibitors (PKIs) have become an effective strategy for cancer patients. However, hepatotoxicity is a major safety concern of these drugs, since the majority are reported to increase transaminases, and few of them (Idelalisib, Lapatinib, Pazopanib, Pexidartinib, Ponatinib, Regorafenib, Sunitinib) have a boxed label warning. The exact rate of PKI-induced hepatoxicity is not well defined due to the fact that the majority of data arise from pre-registration or registration trials on fairly selected patients, and the post-marketing data are often based only on the most severe described cases, whereas most real practice studies do not include drug-related hepatotoxicity as an end point. Although these side effects are usually reversible by dose adjustment or therapy suspension, or by switching to an alternative PKI, and fatality is uncommon, all patients undergoing PKIs should be carefully pre-evaluated and monitored. The management of this complication requires an individually tailored reappraisal of the risk/benefit ratio, especially in patients who are responding to therapy. This review reports the currently available data on the risk and management of hepatotoxicity of all the approved PKIs.
Collapse
Affiliation(s)
- Mauro Viganò
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Correspondence: ; Tel.: +39-035-2674259; Fax: +39-035-2674964
| | - Marta La Milia
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Maria Vittoria Grassini
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Section of Gastroenterology & Hepatology, Department of Health Promotion Sciences Maternal and Infant Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, 90127 Palermo, Italy
| | - Nicola Pugliese
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Massimo De Giorgio
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Stefano Fagiuoli
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Gastroenterology, Department of Medicine, University of Milan Bicocca, 20126 Milan, Italy
| |
Collapse
|
27
|
Role of c-Src and reactive oxygen species in cardiovascular diseases. Mol Genet Genomics 2023; 298:315-328. [PMID: 36700976 DOI: 10.1007/s00438-023-01992-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
Oxidative stress, caused by the over production of oxidants or inactivity of antioxidants, can modulate the redox state of several target proteins such as tyrosine kinases, mitogen-activated protein kinases and tyrosine phosphatases. c-Src is one such non-receptor tyrosine kinase which activates NADPH oxidases (Noxs) in response to various growth factors and shear stress. Interaction between c-Src and Noxs is influenced by cell type and primary messengers such as angiotensin II, which binds to G-protein coupled receptor and activates the intracellular signaling cascade. c-Src stimulated activation of Noxs results in elevated release of intracellular and extracellular reactive oxygen species (ROS). These ROS species disturb vascular homeostasis and cause cardiac hypertrophy, coronary artery disease, atherosclerosis and hypertension. Interaction between c-Src and ROS in the pathobiology of cardiac fibrosis is hypothesized to be influenced by cell type and stimuli. c-Src and ROS have a bidirectional relationship, thus increased ROS levels due to c-Src mediated activation of Noxs can further activate c-Src by promoting the oxidation and sulfenylation of critical cysteine residues. This review highlights the role of c-Src and ROS in mediating downstream signaling pathways underlying cardiovascular diseases. Furthermore, due to the central role of c-Src in activation of various signaling proteins involved in differentiation, migration, proliferation, and cytoskeletal reorganization of vascular cells, it is presented as therapeutic target for treating cardiovascular diseases except cardiac fibrosis.
Collapse
|
28
|
Noguchi R, Yoshimura A, Uchino J, Takeda T, Chihara Y, Ota T, Hiranuma O, Gyotoku H, Takayama K, Kondo T. Comprehensive Kinase Activity Profiling Revealed the Kinase Activity Patterns Associated with the Effects of EGFR Tyrosine Kinase Inhibitor Therapy in Advanced Non-Small-Cell Lung Cancer Patients with Sensitizing EGFR Mutations. Proteomes 2023; 11:proteomes11010006. [PMID: 36810562 PMCID: PMC9944465 DOI: 10.3390/proteomes11010006] [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: 11/08/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 02/08/2023] Open
Abstract
EGFR mutations are strong predictive markers for EGFR tyrosine kinase inhibitor (EGFR-TKI) therapy in patients with non-small-cell lung cancer (NSCLC). Although NSCLC patients with sensitizing EGFR mutations have better prognoses, some patients exhibit worse prognoses. We hypothesized that various activities of kinases could be potential predictive biomarkers for EGFR-TKI treatment among NSCLC patients with sensitizing EGFR mutations. In 18 patients with stage IV NSCLC, EGFR mutations were detected and comprehensive kinase activity profiling was performed using the peptide array PamStation12 for 100 tyrosine kinases. Prognoses were observed prospectively after the administration of EGFR-TKIs. Finally, the kinase profiles were analyzed in combination with the prognoses of the patients. Comprehensive kinase activity analysis identified specific kinase features, consisting of 102 peptides and 35 kinases, in NSCLC patients with sensitizing EGFR mutations. Network analysis revealed seven highly phosphorylated kinases: CTNNB1, CRK, EGFR, ERBB2, PIK3R1, PLCG1, and PTPN11. Pathway analysis and Reactome analysis revealed that the PI3K-AKT and RAF/ MAPK pathways were significantly enriched in the poor prognosis group, being consistent with the outcome of the network analysis. Patients with poor prognoses exhibited high activation of EGFR, PIK3R1, and ERBB2. Comprehensive kinase activity profiles may provide predictive biomarker candidates for screening patients with advanced NSCLC harboring sensitizing EGFR mutations.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Akihiro Yoshimura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Junji Uchino
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Bannan Central Hospital, Shizuoka 438-0814, Japan
| | - Takayuki Takeda
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto 602-8026, Japan
| | - Yusuke Chihara
- Department of Respiratory Medicine, Uji-Tokushukai Medical Center, Kyoto 611-0041, Japan
| | - Takayo Ota
- Department of Medical Oncology, Izumi City General Hospital, Osaka 594-0073, Japan
| | - Osamu Hiranuma
- Department of Respiratory Medicine, Otsu City Hospital, Shiga 520-0804, Japan
| | - Hiroshi Gyotoku
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Correspondence: ; Tel.: +81-3-3542-2511 (ext. 3419)
| |
Collapse
|
29
|
Burnage SC, Bell J, Wan W, Kislenko E, Rurack K. Combining a hybrid chip and tube microfluidic system with fluorescent molecularly imprinted polymer (MIP) core-shell particles for the derivatisation, extraction, and detection of peptides with N-terminating phosphorylated tyrosine. LAB ON A CHIP 2023; 23:466-474. [PMID: 36655759 DOI: 10.1039/d2lc00955b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The reliable identification and quantitation of phosphorylated amino acids, peptides and proteins is one of the key challenges in contemporary bioanalytical research, an area of particular interest when attempting to diagnose and treat diseases at an early stage. We have developed a synthetic probe for targeting phosphorylated amino acids, based on core-shell submicron-sized particles consisting of a silica core, coated with a molecularly imprinted polymer (MIP) shell. The MIP layer contains a fluorescent probe crosslinker which binds selectively to phosphorylated tyrosine (pY) moieties with a significant imprinting factor (IF) and responds with a "light-up" fluorescence signal. The bead-based ratiometric detection scheme has been successfully transferred to a microfluidic chip format and its applicability to rapid assays has been exemplarily shown by discriminating a pY-terminating oligopeptide against its non-phosphorylated counterpart. Such miniaturised devices could lead to an automated pY or pY N-terminated peptide measurement system in the future. The setup combines a modular microfluidic system for amino acid derivatisation, extraction (by micropillar co-flow) and selective adsorption and detection with the fluorescent MIP core-shell particle probes. A miniaturised optical assembly for low-light fluorescence measurements was also developed, based on miniaturised opto-electronic parts and optical fibres. The emission from the MIP particles upon binding of pY or pY N-terminated peptides could be monitored in real-time.
Collapse
Affiliation(s)
- Samual C Burnage
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - Jérémy Bell
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - Wei Wan
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - Evgeniia Kislenko
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - Knut Rurack
- Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| |
Collapse
|
30
|
JAK inhibitors and risk of major cardiovascular events or venous thromboembolism: a self-controlled case series study. Eur J Clin Pharmacol 2022; 78:1981-1990. [DOI: 10.1007/s00228-022-03402-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/07/2022] [Indexed: 11/03/2022]
|
31
|
Sueca-Comes M, Rusu EC, Grabowska AM, Bates DO. Looking Under the Lamppost: The Search for New Cancer Targets in the Human Kinome. Pharmacol Rev 2022; 74:1136-1145. [PMID: 36180110 DOI: 10.1124/pharmrev.121.000410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 02/02/2022] [Accepted: 02/15/2022] [Indexed: 11/22/2022] Open
Abstract
The number of cancer drugs is increasing as new chemical entities are developed to target molecules, often protein kinases, driving cancer progression. In 2009, Fedorov et al. identified that of the protein kinases in the human kinome, most of the focus has been on a small subset. They highlighted that many poorly investigated protein kinases were cancer drivers, but there was no relationship between publications and involvement in cancer development or progression. Since 2009, there has been a doubling in the number of publications, patents, and drugs targeting the kinome. To determine whether this was an expansion in knowledge of well-studied targets-searching in the light under the lamppost-or an explosion of investigations into previously poorly investigated targets, we searched the literature for publications on each kinase, updating Federov et al.'s assessment of the druggable kinome. The proportion of papers focusing on the 50 most-studied kinases had not changed, and the makeup of those 50 had barely changed. The majority of new drugs (80%) were against the same group of 50 kinases identified as targets 10 years ago, and the proportion of studies investigating previously poorly investigated kinases (<1%) was unchanged. With three exceptions [p38 mitogenactivated protein kinase (p38a), AMP-activated protein kinase catalytic α-subunit 1,2, and B-Raf proto-oncogene (BRAF) serine/threonine kinase], >95% of publications addressing kinases still focused on a relatively small proportion (<50%) of the human kinome independently of their involvement as cancer drivers. There is, therefore, still extensive scope for discovery of therapeutics targeting different protein kinases in cancer and still a bias toward well-characterized targets over the innovative searchlight into the unknown. SIGNIFICANCE STATEMENT: This study presents evidence that drug discovery efforts in cancer are still to some extent focused on a narrow group of well-studied kinases 10 years after the identification of multiple novel cancer targets in the human kinome. This suggests that there is still room for researchers in academia, industry, and the not-for-profit sector to develop new and diverse therapies targeting kinases for cancer.
Collapse
Affiliation(s)
- Mireia Sueca-Comes
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| | - Elena Cristina Rusu
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| | - Anna M Grabowska
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| | - David O Bates
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| |
Collapse
|
32
|
Zhang Q, Kounde CS, Mondal M, Greenfield JL, Baker JR, Kotelnikov S, Ignatov M, Tinworth CP, Zhang L, Conole D, De Vita E, Kozakov D, McCluskey A, Harling JD, Fuchter MJ, Tate EW. Light-mediated multi-target protein degradation using arylazopyrazole photoswitchable PROTACs (AP-PROTACs). Chem Commun (Camb) 2022; 58:10933-10936. [PMID: 36065962 PMCID: PMC9521323 DOI: 10.1039/d2cc03092f] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Light-activable spatiotemporal control of PROTAC-induced protein degradation was achieved with novel arylazopyrazole photoswitchable PROTACs (AP-PROTACs). The use of a promiscuous kinase inhibitor in the design enables this unique photoswitchable PROTAC to selectively degrade four protein kinases together with on/off optical control using different wavelengths of light. A new class of arylazopyrazole photoswitchable PROTACs (AP-PROTACs) enables light-triggered degradation of a specific ensemble of protein kinases.![]()
Collapse
Affiliation(s)
- Qisi Zhang
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Cyrille S Kounde
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Milon Mondal
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Jake L Greenfield
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Jennifer R Baker
- Chemistry, School of Environmental & Life Sciences, the University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Sergei Kotelnikov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA.,Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Mikhail Ignatov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA.,Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Christopher P Tinworth
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Leran Zhang
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Daniel Conole
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Elena De Vita
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA.,Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, the University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - John D Harling
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Matthew J Fuchter
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK.
| |
Collapse
|
33
|
Lorusso D, Danesi R, Locati LD, Masi G, De Giorgi U, Gadducci A, Pignata S, Sabbatini R, Savarese A, Valabrega G, Zamagni C, Colombo N. Optimizing the use of lenvatinib in combination with pembrolizumab in patients with advanced endometrial carcinoma. Front Oncol 2022; 12:979519. [PMID: 36212444 PMCID: PMC9535356 DOI: 10.3389/fonc.2022.979519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction The combination of lenvatinib plus pembrolizumab demonstrated a relevant clinical benefit in patients with endometrial carcinoma. The safety profile was consistent with the established profiles of each drug in monotherapy, with the most frequent adverse events being hypertension, an on-target effect, hypothyroidism, diarrhea, nausea, vomiting, loss of appetite, fatigue, and weight loss. Areas covered We first review the rationale based on the combination of a VEGFR inhibitor and an immune checkpoint inhibitor, highlighting the main pharmacokinetic and pharmacodynamic features of lenvatinib. Next, we focus on the common adverse events associated with lenvatinib and guide how to optimally prevent, detect, and manage them, while minimizing interruptions during lenvatinib treatment. Discussion The side effects profile of lenvatinib is very well known, being similar across different tumor types. Most toxicities can be preventable. An appropriate, proactive, and thorough management of lenvatinib toxicities during treatment is required to maximize potential lenvatinib efficacy. Adverse events should be detected as early as possible, by both carefully monitoring the patient from lenvatinib initiation and preventing their occurrence. Patients should be followed also during treatment as some adverse events, e.g., cardiac dysfunction might appear later. Increased awareness on risk to benefit ratio among clinicians would be helpful to avoid dose interruptions or discontinuation of lenvatinib, with preferring other medical interventions and supportive care.
Collapse
Affiliation(s)
- Domenica Lorusso
- Department of Clinical Research Planning, Fondazione Policlinico Universitario A Gemelli Istituto di Ricerca e Cura a carattere scientifico (IRCCS), Rome, Italy
- Department of Life Science and Public Health, Catholic University of Sacred Heart, Rome, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Deborah Locati
- Translational Oncology Unit, Istituto di Ricerca e Cura a carattere scientifico (IRCCS) Istituti Clinici Scientifici (ICS) Maugeri, Pavia, Italy
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto di Ricerca e Cura a carattere scientifico (IRCCS) Istituto Romagnolo per lo Studio dei Tumori (IRST), Dino Amadori, Meldola, Italy
| | - Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Pisa, Italy
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori Istituto di Ricerca e Cura a carattere scientifico (IRCCS) “Fondazione Giovanni Pascale”, Naples, Italy
| | - Roberto Sabbatini
- Department of Urology and Gynecology, Istituto Nazionale Tumori Istituto di Ricerca e Cura a carattere scientifico (IRCCS) “Fondazione Giovanni Pascale”, Naples, Italy
| | - Antonella Savarese
- Division of Medical Oncology 1, Istituto di Ricerca e Cura a carattere scientifico (IRCCS) -Regina Elena National Cancer Institute, Rome, Italy
| | - Giorgio Valabrega
- University of Torino-Struttura Complessa a Direzione Universitaria (S.C.D.U.) Oncologia Azienda Ospedaliera (A.O) Ordine Mauriziano-Ospedale Umberto I, Torino, Italy
| | - Claudio Zamagni
- Addarii Medical Oncology, Istituto di Ricerca e Cura a carattere scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Nicoletta Colombo
- School of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
- Department of Oncological Gynecology, European Institute of Oncology (IEO) Istituto di Ricerca e Cura a carattere scientifico (IRCCS), Milan, Italy
| |
Collapse
|
34
|
Dewangan PS, Beraki TG, Paiz EA, Appiah Mensah D, Chen Z, Reese ML. Divergent kinase WNG1 is regulated by phosphorylation of an atypical activation sub-domain. Biochem J 2022; 479:1877-1889. [PMID: 35938919 PMCID: PMC9555795 DOI: 10.1042/bcj20220076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
Apicomplexan parasites like Toxoplasma gondii grow and replicate within a specialized organelle called the parasitophorous vacuole. The vacuole is decorated with parasite proteins that integrate into the membrane after trafficking through the parasite secretory system as soluble, chaperoned complexes. A regulator of this process is an atypical protein kinase called WNG1. Phosphorylation by WNG1 appears to serve as a switch for membrane integration. However, like its substrates, WNG1 is secreted from the parasite dense granules, and its activity must, therefore, be tightly regulated until the correct membrane is encountered. Here, we demonstrate that, while another member of the WNG family can adopt multiple multimeric states, WNG1 is monomeric and therefore not regulated by multimerization. Instead, we identify two phosphosites on WNG1 that are required for its kinase activity. Using a combination of in vitro biochemistry and structural modeling, we identify basic residues that are also required for WNG1 activity and appear to recognize the activating phosphosites. Among these coordinating residues are the 'HRD' Arg, which recognizes activation loop phosphorylation in canonical kinases. WNG1, however, is not phosphorylated on its activation loop, but rather on atypical phosphosites on its C-lobe. We propose a simple model in which WNG1 is activated by increasing ATP concentration above a critical threshold once the kinase traffics to the parasitophorous vacuole.
Collapse
Affiliation(s)
- Pravin S. Dewangan
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
| | - Tsebaot G. Beraki
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
| | - E. Ariana Paiz
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
| | - Delia Appiah Mensah
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
- Honors College, University of Texas at Dallas, Richardson, TX, U.S.A
| | - Zhe Chen
- Department of Biophysics, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
| | - Michael L. Reese
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
- Department of Biochemistry, University of Texas, Southwestern Medical Center, Dallas, TX, U.S.A
| |
Collapse
|
35
|
Wei S, Zhang J, Shi R, Yu Z, Chen X, Wang H. Identification of an integrated kinase-related prognostic gene signature associated with tumor immune microenvironment in human uterine corpus endometrial carcinoma. Front Oncol 2022; 12:944000. [PMID: 36158685 PMCID: PMC9491090 DOI: 10.3389/fonc.2022.944000] [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: 05/14/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
In the worldwide, uterine corpus endometrial carcinoma (UCEC) is the sixth most common malignancy in women, and the number of women diagnosed is increasing. Kinase plays an important role in the occurrence and development of malignant tumors. However, the research about kinase in endometrial cancer is still unclear. Here, we first downloaded the gene expression data of 552 UCEC patients and 23 healthy endometrial tissues from The Cancer Genome Atlas (TCGA), obtained 538 kinase-related genes from the previous literature, and calculated 67 differentially expressed kinases. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were referenced to identify multiple important biological functions and signaling pathways related to 67 differentially expressed kinases. Using univariate Cox regression and Least absolute shrinkage and selection operator (LASSO), seven kinases (ALPK2, CAMKV, TTK, PTK6, MAST1, CIT, and FAM198B) were identified to establish a prognostic model of endometrial cancer. Then, patients were divided into high- and low-risk groups based on risk scores. Receiver operating characteristic (ROC) curves were plotted to evaluate that the model had a favorable predictive ability. Kaplan–Meier survival analysis suggested that high-risk groups experienced worse overall survival than low-risk groups. qRT-PCR and ISH assays confirmed the consistency between predicted candidate genes and real sample contents. CIBERSORT algorithm and ssGSEA were adopted to investigate the relationship between this signature and tumor immune microenvironment, and revealed that in low- and high-risk groups, the types of tumor-infiltrating immune cells and the immune cell-related functions were significantly different. In summary, a seven-gene signature risk model has been constructed, and could accurately predict the prognosis of UCEC, which may offer ideas and breakthrough points to the kinase-associated development of UCEC.
Collapse
Affiliation(s)
- Sitian Wei
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Shi
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhicheng Yu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingwei Chen
- Department of Industrial engineering, Tsinghua University, Beijing, China
| | - Hongbo Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hongbo Wang,
| |
Collapse
|
36
|
Rudloff U. Emerging kinase inhibitors for the treatment of pancreatic ductal adenocarcinoma. Expert Opin Emerg Drugs 2022; 27:345-368. [PMID: 36250721 PMCID: PMC9793333 DOI: 10.1080/14728214.2022.2134346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/22/2022] [Accepted: 10/06/2022] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Pancreatic cancer is one of the deadliest solid organ cancers. In the absence of specific warning symptoms pancreatic cancer is diagnosed notoriously late. Current systemic chemotherapy regimens extend survival by a mere few months. With the advances in genetic, proteomic, and immunological profiling there is strong rationale to test kinase inhibitors to improve outcome. AREAS COVERED This review article provides a comprehensive summary of approved treatments and past, present, and future developments of kinase inhibitors in pancreatic cancer. Emerging roles of protein kinase inhibitors are discussed in the context of the unique stroma, the lack of high-prevalence therapeutic targets and rapid emergence of acquired resistance, novel immuno-oncology kinase targets, and recent medicinal chemistry advances. EXPERT OPINION Due to the to-date frequent failure of protein kinase inhibitors indiscriminately administered to unselected pancreatic cancer patients, there is a shift toward the development of these agents in molecularly defined subgroups which are more likely to respond. The development of accurate biomarkers to select patients who are the best candidates based on a detailed understanding of mechanism of action, pro-survival roles, and mediation of resistance of targeted kinases will be critical for the future development of protein kinase inhibitors in this disease.
Collapse
Affiliation(s)
- Udo Rudloff
- Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
37
|
Deboever E, Fistrovich A, Hulme C, Dunckley T. The Omnipresence of DYRK1A in Human Diseases. Int J Mol Sci 2022; 23:ijms23169355. [PMID: 36012629 PMCID: PMC9408930 DOI: 10.3390/ijms23169355] [Citation(s) in RCA: 18] [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: 07/07/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023] Open
Abstract
The increasing population will challenge healthcare, particularly because the worldwide population has never been older. Therapeutic solutions to age-related disease will be increasingly critical. Kinases are key regulators of human health and represent promising therapeutic targets for novel drug candidates. The dual-specificity tyrosine-regulated kinase (DYRKs) family is of particular interest and, among them, DYRK1A has been implicated ubiquitously in varied human diseases. Herein, we focus on the characteristics of DYRK1A, its regulation and functional role in different human diseases, which leads us to an overview of future research on this protein of promising therapeutic potential.
Collapse
Affiliation(s)
- Estelle Deboever
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Correspondence: (E.D.); (T.D.)
| | - Alessandra Fistrovich
- Department of Chemistry and Biochemistry, College of Science, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA
| | - Christopher Hulme
- Department of Chemistry and Biochemistry, College of Science, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA
| | - Travis Dunckley
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Correspondence: (E.D.); (T.D.)
| |
Collapse
|
38
|
Abstract
The nitrogen mustards are powerful cytotoxic and lymphoablative agents and have been used for more than 60 years. They are employed in the treatment of cancers, sarcomas, and hematologic malignancies. Cyclophosphamide, the most versatile of the nitrogen mustards, also has a place in stem cell transplantation and the therapy of autoimmune diseases. Adverse effects caused by the nitrogen mustards on the central nervous system, kidney, heart, bladder, and gonads remain important issues. Advances in analytical techniques have facilitated the investigation of the pharmacokinetics of the nitrogen mustards, especially the oxazaphosphorines, which are prodrugs requiring metabolic activation. Enzymes involved in the metabolism of cyclophosphamide and ifosfamide are very polymorphic, but a greater understanding of the pharmacogenomic influences on their activity has not yet translated into a personalized medicine approach. In addition to damaging DNA, the nitrogen mustards can act through other mechanisms, such as antiangiogenesis and immunomodulation. The immunomodulatory properties of cyclophosphamide are an area of current exploration. In particular, cyclophosphamide decreases the number and activity of regulatory T cells, and the interaction between cyclophosphamide and the intestinal microbiome is now recognized as an important factor. New derivatives of the nitrogen mustards continue to be assessed. Oxazaphosphorine analogs have been synthesized in attempts to both improve efficacy and reduce toxicity, with varying degrees of success. Combinations of the nitrogen mustards with monoclonal antibodies and small-molecule targeted agents are being evaluated. SIGNIFICANCE STATEMENT: The nitrogen mustards are important, well-established therapeutic agents that are used to treat a variety of diseases. Their role is continuing to evolve.
Collapse
Affiliation(s)
- Martin S Highley
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Bart Landuyt
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Hans Prenen
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Peter G Harper
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Ernst A De Bruijn
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| |
Collapse
|
39
|
Gyenis L, Menyhart D, Cruise ES, Jurcic K, Roffey SE, Chai DB, Trifoi F, Fess SR, Desormeaux PJ, Núñez de Villavicencio Díaz T, Rabalski AJ, Zukowski SA, Turowec JP, Pittock P, Lajoie G, Litchfield DW. Chemical Genetic Validation of CSNK2 Substrates Using an Inhibitor-Resistant Mutant in Combination with Triple SILAC Quantitative Phosphoproteomics. Front Mol Biosci 2022; 9:909711. [PMID: 35755813 PMCID: PMC9225150 DOI: 10.3389/fmolb.2022.909711] [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: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
Casein Kinase 2 (CSNK2) is an extremely pleiotropic, ubiquitously expressed protein kinase involved in the regulation of numerous key biological processes. Mapping the CSNK2-dependent phosphoproteome is necessary for better characterization of its fundamental role in cellular signalling. While ATP-competitive inhibitors have enabled the identification of many putative kinase substrates, compounds targeting the highly conserved ATP-binding pocket often exhibit off-target effects limiting their utility for definitive kinase-substrate assignment. To overcome this limitation, we devised a strategy combining chemical genetics and quantitative phosphoproteomics to identify and validate CSNK2 substrates. We engineered U2OS cells expressing exogenous wild type CSNK2A1 (WT) or a triple mutant (TM, V66A/H160D/I174A) with substitutions at residues important for inhibitor binding. These cells were treated with CX-4945, a clinical-stage inhibitor of CSNK2, and analyzed using large-scale triple SILAC (Stable Isotope Labelling of Amino Acids in Cell Culture) quantitative phosphoproteomics. In contrast to wild-type CSNK2A1, CSNK2A1-TM retained activity in the presence of CX-4945 enabling identification and validation of several CSNK2 substrates on the basis of their increased phosphorylation in cells expressing CSNK2A1-TM. Based on high conservation within the kinase family, we expect that this strategy can be broadly adapted for identification of other kinase-substrate relationships.
Collapse
Affiliation(s)
- Laszlo Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Daniel Menyhart
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Edward S Cruise
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Kristina Jurcic
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Scott E Roffey
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Darren B Chai
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Flaviu Trifoi
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Sam R Fess
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Paul J Desormeaux
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | | | - Adam J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Stephanie A Zukowski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Jacob P Turowec
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Paula Pittock
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Gilles Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - David W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada.,Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
40
|
Luxen M, van Meurs M, Molema G. Unlocking the Untapped Potential of Endothelial Kinase and Phosphatase Involvement in Sepsis for Drug Treatment Design. Front Immunol 2022; 13:867625. [PMID: 35634305 PMCID: PMC9136877 DOI: 10.3389/fimmu.2022.867625] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Sepsis is a devastating clinical condition that can lead to multiple organ failure and death. Despite advancements in our understanding of molecular mechanisms underlying sepsis and sepsis-associated multiple organ failure, no effective therapeutic treatment to directly counteract it has yet been established. The endothelium is considered to play an important role in sepsis. This review highlights a number of signal transduction pathways involved in endothelial inflammatory activation and dysregulated endothelial barrier function in response to sepsis conditions. Within these pathways – NF-κB, Rac1/RhoA GTPases, AP-1, APC/S1P, Angpt/Tie2, and VEGF/VEGFR2 – we focus on the role of kinases and phosphatases as potential druggable targets for therapeutic intervention. Animal studies and clinical trials that have been conducted for this purpose are discussed, highlighting reasons why they might not have resulted in the expected outcomes, and which lessons can be learned from this. Lastly, opportunities and challenges that sepsis and sepsis-associated multiple organ failure research are currently facing are presented, including recommendations on improved experimental design to increase the translational power of preclinical research to the clinic.
Collapse
Affiliation(s)
- Matthijs Luxen
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Matthijs Luxen,
| | - Matijs van Meurs
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Grietje Molema
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
41
|
Harris JA, Fairweather E, Byrne DP, Eyers PA. Analysis of human Tribbles 2 (TRIB2) pseudokinase. Methods Enzymol 2022; 667:79-99. [PMID: 35525562 DOI: 10.1016/bs.mie.2022.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human Tribbles 2 (TRIB2) is a cancer-associated pseudokinase with a broad human protein interactome, including the well-studied AKT, C/EBPα and MAPK modules. Several lines of evidence indicate that human TRIB2 promotes cell survival and drug-resistance in solid tumors and blood cancers and is therefore of interest as a potential therapeutic target, although its physiological functions remain relatively poorly understood. The unique TRIB2 pseudokinase domain lacks the canonical 'DFG' motif, and subsequently possesses very low affinity for ATP in both the presence and absence of metal ions. However, TRIB2 also contains a unique cysteine-rich αC-helix, which interacts with a conserved peptide motif in its own carboxyl-terminal tail. This regulatory flanking region drives regulated interactions with distinct E3 ubiquitin ligases that serve to control the stability and turnover of TRIB2 client proteins. TRIB2 is also a low-affinity target of several known small-molecule protein kinase inhibitors, which were originally identified using purified recombinant TRIB2 proteins and a thermal shift assay. In this chapter, we discuss laboratory-based procedures for purification, stabilization and analysis of human TRIB2, including screening procedures that can be used for the identification of both reversible and covalent small molecule ligands.
Collapse
Affiliation(s)
- John A Harris
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emma Fairweather
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Dominic P Byrne
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Patrick A Eyers
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
42
|
Kirchoff KE, Gomez SM. EMBER: multi-label prediction of kinase-substrate phosphorylation events through deep learning. Bioinformatics 2022; 38:2119-2126. [PMID: 35157015 PMCID: PMC9004653 DOI: 10.1093/bioinformatics/btac083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 12/09/2021] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
MOTIVATION Kinase-catalyzed phosphorylation of proteins forms the backbone of signal transduction within the cell, enabling the coordination of numerous processes such as the cell cycle, apoptosis, and differentiation. Although on the order of 105 phosphorylation events have been described, we know the specific kinase performing these functions for <5% of cases. The ability to predict which kinases initiate specific individual phosphorylation events has the potential to greatly enhance the design of downstream experimental studies, while simultaneously creating a preliminary map of the broader phosphorylation network that controls cellular signaling. RESULTS We describe Embedding-based multi-label prediction of phosphorylation events (EMBER), a deep learning method that integrates kinase phylogenetic information and motif-dissimilarity information into a multi-label classification model for the prediction of kinase-motif phosphorylation events. Unlike previous deep learning methods that perform single-label classification, we restate the task of kinase-motif phosphorylation prediction as a multi-label problem, allowing us to train a single unified model rather than a separate model for each of the 134 kinase families. We utilize a Siamese neural network to generate novel vector representations, or an embedding, of peptide motif sequences, and we compare our novel embedding to a previously proposed peptide embedding. Our motif vector representations are used, along with one-hot encoded motif sequences, as input to a classification neural network while also leveraging kinase phylogenetic relationships into our model via a kinase phylogeny-weighted loss function. Results suggest that this approach holds significant promise for improving the known map of phosphorylation relationships that underlie kinome signaling. AVAILABILITY AND IMPLEMENTATION The data and code underlying this article are available in a GitHub repository at https://github.com/gomezlab/EMBER. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Kathryn E Kirchoff
- Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shawn M Gomez
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- North Carolina State University, Raleigh, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
43
|
Shining Light on Protein Kinase Biomarkers with Fluorescent Peptide Biosensors. Life (Basel) 2022; 12:life12040516. [PMID: 35455007 PMCID: PMC9026840 DOI: 10.3390/life12040516] [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: 03/01/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 11/23/2022] Open
Abstract
Protein kinases (PKs) are established gameplayers in biological signalling pathways, and a large body of evidence points to their dysregulation in diseases, in particular cancer, where rewiring of PK networks occurs frequently. Fluorescent biosensors constitute attractive tools for probing biomolecules and monitoring dynamic processes in complex samples. A wide variety of genetically encoded and synthetic biosensors have been tailored to report on PK activities over the last decade, enabling interrogation of their function and insight into their behaviour in physiopathological settings. These optical tools can further be used to highlight enzymatic alterations associated with the disease, thereby providing precious functional information which cannot be obtained through conventional genetic, transcriptomic or proteomic approaches. This review focuses on fluorescent peptide biosensors, recent developments and strategies that make them attractive tools to profile PK activities for biomedical and diagnostic purposes, as well as insights into the challenges and opportunities brought by this unique toolbox of chemical probes.
Collapse
|
44
|
Demuro S, Sauvey C, Tripathi SK, Di Martino RMC, Shi D, Ortega JA, Russo D, Balboni B, Giabbai B, Storici P, Girotto S, Abagyan R, Cavalli A. ARN25068, a versatile starting point towards triple GSK-3β/FYN/DYRK1A inhibitors to tackle tau-related neurological disorders. Eur J Med Chem 2022; 229:114054. [PMID: 34959172 PMCID: PMC9704499 DOI: 10.1016/j.ejmech.2021.114054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
The human kinome plays a crucial role in several pathways. Its dysregulation has been linked to diverse central nervous system (CNS)-related disorders with a drastic impact on the aging population. Among them, tauopathies, such as Alzheimer's Disease (AD) and Frontotemporal Lobar degeneration (FTLD-tau), are neurodegenerative disorders pathologically defined by the presence of hyperphosphorylated tau-positive intracellular inclusions known as neurofibrillary tangles (NFTs). Compelling evidence has reported the great potential of the simultaneous modulation of multiple protein kinases (PKs) involved in abnormal tau phosphorylation through a concerted pharmacological approach to achieve a superior therapeutic effect relative to classic "one target, one drug" approaches. Here, we report on the identification and characterization of ARN25068 (4), a low nanomolar and well-balanced dual GSK-3β and FYN inhibitor, which also shows inhibitory activity against DYRK1A, an emerging target in AD and tauopathies. Computational and X-Ray studies highlight compound 4's typical H-bonding pattern of ATP-competitive inhibitors at the binding sites of all three PKs. In a tau phosphorylation assay on Tau0N4R-TM-tGFP U2OS cell line, 4 reduces the extent of tau phosphorylation, promoting tau-stabilized microtubule bundles. In conclusion, this compound emerges as a promising prototype for further SAR explorations to develop potent and well-balanced triple GSK-3β/FYN/DYRK1A inhibitors to tackle tau hyperphosphorylation.
Collapse
Affiliation(s)
- Stefania Demuro
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy; Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Conall Sauvey
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Shailesh K Tripathi
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - Rita M C Di Martino
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Jose A Ortega
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - Debora Russo
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Beatrice Balboni
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy; Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Barbara Giabbai
- Protein Facility, Elettra Sincrotrone Trieste S.C.p.A., SS 14 - Km 163, 5 in AREA Science Park, 34149, Trieste, Italy
| | - Paola Storici
- Protein Facility, Elettra Sincrotrone Trieste S.C.p.A., SS 14 - Km 163, 5 in AREA Science Park, 34149, Trieste, Italy
| | - Stefania Girotto
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, United States.
| | - Andrea Cavalli
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy; Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy.
| |
Collapse
|
45
|
Byrne DP, Omar MH, Kennedy EJ, Eyers PA, Scott JD. Biochemical Analysis of AKAP-Anchored PKA Signaling Complexes. Methods Mol Biol 2022; 2483:297-317. [PMID: 35286684 PMCID: PMC9518671 DOI: 10.1007/978-1-0716-2245-2_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Generation of the prototypic second messenger cAMP instigates numerous signaling events. A major intracellular target of cAMP is Protein kinase A (PKA), a Ser/Thr protein kinase. Where and when this enzyme is activated inside the cell has profound implications on the functional impact of PKA. It is now well established that PKA signaling is focused locally into subcellular signaling "islands" or "signalosomes." The A-Kinase Anchoring Proteins (AKAPs) play a critical role in this process by dictating spatial and temporal aspects of PKA action. Genetically encoded biosensors, small molecule and peptide-based disruptors of PKA signaling are valuable tools for rigorous investigation of local PKA action at the biochemical level. This chapter focuses on approaches to evaluate PKA signaling islands, including a simple assay for monitoring the interaction of an AKAP with a tunable PKA holoenzyme. The latter approach evaluates the composition of PKA holoenzymes, in which regulatory subunits and catalytic subunits can be visualized in the presence of test compounds and small-molecule inhibitors.
Collapse
Affiliation(s)
- Dominic P Byrne
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Liverpool, UK
| | - Mitchell H Omar
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Patrick A Eyers
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Liverpool, UK.
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
46
|
Tahtouh T, Durieu E, Villiers B, Bruyère C, Nguyen TL, Fant X, Ahn KH, Khurana L, Deau E, Lindberg MF, Sévère E, Miege F, Roche D, Limanton E, L'Helgoual'ch JM, Burgy G, Guiheneuf S, Herault Y, Kendall DA, Carreaux F, Bazureau JP, Meijer L. Structure-Activity Relationship in the Leucettine Family of Kinase Inhibitors. J Med Chem 2021; 65:1396-1417. [PMID: 34928152 DOI: 10.1021/acs.jmedchem.1c01141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The protein kinase DYRK1A is involved in Alzheimer's disease, Down syndrome, diabetes, viral infections, and leukemia. Leucettines, a family of 2-aminoimidazolin-4-ones derived from the marine sponge alkaloid Leucettamine B, have been developed as pharmacological inhibitors of DYRKs (dual specificity, tyrosine phosphorylation regulated kinases) and CLKs (cdc2-like kinases). We report here on the synthesis and structure-activity relationship (SAR) of 68 Leucettines. Leucettines were tested on 11 purified kinases and in 5 cellular assays: (1) CLK1 pre-mRNA splicing, (2) Threonine-212-Tau phosphorylation, (3) glutamate-induced cell death, (4) autophagy and (5) antagonism of ligand-activated cannabinoid receptor CB1. The Leucettine SAR observed for DYRK1A is essentially identical for CLK1, CLK4, DYRK1B, and DYRK2. DYRK3 and CLK3 are less sensitive to Leucettines. In contrast, the cellular SAR highlights correlations between inhibition of specific kinase targets and some but not all cellular effects. Leucettines deserve further development as potential therapeutics against various diseases on the basis of their molecular targets and cellular effects.
Collapse
Affiliation(s)
- Tania Tahtouh
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,CNRS, 'Protein Phosphorylation and Human Disease' Group, Station Biologique De Roscoff, Place G. Teissier, Bp 74, 29682 Roscoff, Bretagne, France.,College Of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Emilie Durieu
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,CNRS, 'Protein Phosphorylation and Human Disease' Group, Station Biologique De Roscoff, Place G. Teissier, Bp 74, 29682 Roscoff, Bretagne, France
| | - Benoît Villiers
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Céline Bruyère
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Thu Lan Nguyen
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,Institut De Génétique Et De Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Université de Strasbourg, CNRS UMR7104 & INSERM U964, 67400 Illkirch, France.,Laboratory of Molecular & Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, New York 10021-6399, United States
| | - Xavier Fant
- CNRS, 'Protein Phosphorylation and Human Disease' Group, Station Biologique De Roscoff, Place G. Teissier, Bp 74, 29682 Roscoff, Bretagne, France
| | - Kwang H Ahn
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, Connecticut 06269, United States
| | - Leepakshi Khurana
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, Connecticut 06269, United States
| | - Emmanuel Deau
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Mattias F Lindberg
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Elodie Sévère
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Frédéric Miege
- Edelris, Bâtiment Bioserra 1, 60 avenue Rockefeller, 69008 Lyon, France
| | - Didier Roche
- Edelris, Bâtiment Bioserra 1, 60 avenue Rockefeller, 69008 Lyon, France
| | - Emmanuelle Limanton
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Jean-Martial L'Helgoual'ch
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Guillaume Burgy
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France.,Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Solène Guiheneuf
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Yann Herault
- Institut De Génétique Et De Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Université de Strasbourg, CNRS UMR7104 & INSERM U964, 67400 Illkirch, France
| | - Debra A Kendall
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Storrs, Connecticut 06269, United States
| | - François Carreaux
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Jean-Pierre Bazureau
- Institut des Sciences Chimiques de Rennes ISCR-UMR CNRS 6226, Université de Rennes 1, Campus de Beaulieu, Bât. 10A, CS 74205, 263 Avenue du Général Leclerc, 35042 Rennes Cedex, France
| | - Laurent Meijer
- Manros Therapeutics & Perha Pharmaceuticals, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| |
Collapse
|
47
|
Sergienko NM, Donner DG, Delbridge LMD, McMullen JR, Weeks KL. Protein phosphatase 2A in the healthy and failing heart: New insights and therapeutic opportunities. Cell Signal 2021; 91:110213. [PMID: 34902541 DOI: 10.1016/j.cellsig.2021.110213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
Protein phosphatases have emerged as critical regulators of phosphoprotein homeostasis in settings of health and disease. Protein phosphatase 2A (PP2A) encompasses a large subfamily of enzymes that remove phosphate groups from serine/threonine residues within phosphoproteins. The heterogeneity in PP2A structure, which arises from the grouping of different catalytic, scaffolding and regulatory subunit isoforms, creates distinct populations of catalytically active enzymes (i.e. holoenzymes) that localise to different parts of the cell. This structural complexity, combined with other regulatory mechanisms, such as interaction of PP2A heterotrimers with accessory proteins and post-translational modification of the catalytic and/or regulatory subunits, enables PP2A holoenzymes to target phosphoprotein substrates in a highly specific manner. In this review, we summarise the roles of PP2A in cardiac physiology and disease. PP2A modulates numerous processes that are vital for heart function including calcium handling, contractility, β-adrenergic signalling, metabolism and transcription. Dysregulation of PP2A has been observed in human cardiac disease settings, including heart failure and atrial fibrillation. Efforts are underway, particularly in the cancer field, to develop therapeutics targeting PP2A activity. The development of small molecule activators of PP2A (SMAPs) and other compounds that selectively target specific PP2A holoenzymes (e.g. PP2A/B56α and PP2A/B56ε) will improve understanding of the function of different PP2A species in the heart, and may lead to the development of therapeutics for normalising aberrant protein phosphorylation in settings of cardiac remodelling and dysfunction.
Collapse
Affiliation(s)
- Nicola M Sergienko
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Central Clinical School, Monash University, Clayton VIC 3800, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Clayton VIC 3800, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora VIC 3086, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| |
Collapse
|
48
|
Maillard M, Louveau B, Vilquin P, Goldwirt L, Thomas F, Mourah S. Pharmacogenomics in solid cancers and hematologic malignancies: Improving personalized drug prescription. Therapie 2021; 77:171-183. [PMID: 34922740 DOI: 10.1016/j.therap.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022]
Abstract
The discovery of molecular alterations involved in oncogenesis is evolving rapidly and has led to the development of new innovative targeted therapies in oncology. High-throughput sequencing techniques help to identify genomic targets and to provide predictive molecular biomarkers of response to guide alternative therapeutic strategies. Besides the emergence of these theranostic markers for the new targeted treatments, pharmacogenetic markers (corresponding to genetic variants existing in the constitutional DNA, i.e., the host genome) can help to optimize the use of chemotherapy. In this review, we present the current clinical applications of constitutional PG and the recent concepts and advances in pharmacogenomics, a rapidly evolving field that focuses on various molecular alterations identified on constitutional or somatic (tumor) genome.
Collapse
Affiliation(s)
- Maud Maillard
- Institut Claudius-Regaud, Institut universitaire du cancer de Toulouse, IUCT-Oncopole, 31059 Toulouse, France; Centre de recherches en cancérologie de Toulouse CRCT, 31037 Toulouse, France; Université Paul-Sabatier Toulouse III, 31062 Toulouse, France
| | - Baptiste Louveau
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Paul Vilquin
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Lauriane Goldwirt
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Fabienne Thomas
- Institut Claudius-Regaud, Institut universitaire du cancer de Toulouse, IUCT-Oncopole, 31059 Toulouse, France; Centre de recherches en cancérologie de Toulouse CRCT, 31037 Toulouse, France; Université Paul-Sabatier Toulouse III, 31062 Toulouse, France
| | - Samia Mourah
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France.
| |
Collapse
|
49
|
Krishna AP, John S, Shinde PL, Mishra R. Proteo-transcriptomics meta-analysis identifies SUMO2 as a promising target in glioblastoma multiforme therapeutics. Cancer Cell Int 2021; 21:575. [PMID: 34715855 PMCID: PMC8555349 DOI: 10.1186/s12935-021-02279-y] [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: 07/13/2021] [Accepted: 10/18/2021] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a deadly brain tumour with minimal survival rates due to the ever-expanding heterogeneity, chemo and radioresistance. Kinases are known to crucially drive GBM pathology; however, a rationale therapeutic combination that can simultaneously inhibit multiple kinases has not yet emerged successfully. Results Here, we analyzed the GBM patient data from several publicly available repositories and deduced hub GBM kinases, most of which were identified to be SUMOylated by SUMO2/3 isoforms. Not only the hub kinases but a significant proportion of GBM upregulated genes involved in proliferation, metastasis, invasion, epithelial-mesenchymal transition, stemness, DNA repair, stromal and macrophages maintenance were also identified to be the targets of SUMO2 isoform. Correlatively, high expression of SUMO2 isoform was found to be significantly associated with poor patient survival. Conclusions Although many natural products and drugs are evidenced to target general SUMOylation, however, our meta-analysis strongly calls for the need to design SUMO2/3 or even better SUMO2 specific inhibitors and also explore the SUMO2 transcription inhibitors for universally potential, physiologically non-toxic anti-GBM drug therapy. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02279-y. The major highlights of this study are as follows:Key upregulated hub kinases and coding genes in GBM are found to be targets of SUMO2 conjugation. SUMO2 is significantly expressed in adult primary and recurrent GBMs as well as in pediatric GBM tumours. Orthotropic xenografts from adult and pediatric GBMs confirm high expression of SUMO2 in GBM tumour samples. SUMO2 is significantly associated with patient survival plot and pan-cancer cell fitness. Rationale design of SUMO2 inhibitors or search for its transcriptional inhibitors is urgently required through industry-academia collaboration for an anti-GBM and potentially pan-cancer therapeutics.
Collapse
Affiliation(s)
- Aswani P Krishna
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Sebastian John
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Puja Laxmanrao Shinde
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Rashmi Mishra
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India.
| |
Collapse
|
50
|
Yano S, Kawaoka T, Johira Y, Miura R, Kosaka M, Shirane Y, Murakami S, Amioka K, Naruto K, Ando Y, Kosaka Y, Yamaoka K, Kodama K, Uchikawa S, Fujino H, Ohno A, Nakahara T, Murakami E, Okamoto W, Yamauchi M, Imamura M, Mori K, Arihiro K, Kuroda S, Kobayashi T, Ohdan H, Aikata H. Advanced hepatocellular carcinoma with response to lenvatinib after atezolizumab plus bevacizumab. Medicine (Baltimore) 2021; 100:e27576. [PMID: 34678902 PMCID: PMC8542139 DOI: 10.1097/md.0000000000027576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/07/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Various treatments are available for treating hepatocellular carcinoma (HCC). The immune checkpoint inhibitor combination of atezolizumab plus bevacizumab was recently approved for the treatment of unresectable HCC, but there are few reports on the failure of the combination treatment. Here, we present a case of unresectable HCC with adrenal metastasis that was eventually operated on after lenvatinib (LEN) treatment that followed failed treatment with atezolizumab plus bevacizumab. PATIENT CONCERNS A 68-year-old man was diagnosed with non-alcoholic steatohepatitis-based HCC with adrenal metastasis. DIAGNOSIS Cirrhosis was classified as Child-Pugh score of 5. HCC was diagnosed as Barcelona Clinic Liver Cancer stage C. INTERVENTIONS We initiated treatment with atezolizumab plus bevacizumab. Liver dysfunction appeared 2 days after the first administration but was improved by intravenous rehydration and did not appear after the second course. The HCC shrank, but the adrenal metastasis grew bigger after the fourth course, so we changed the therapy to LEN. After HCC and adrenal metastasis were necrotic by LEN, conversion surgery was performed. OUTCOMES After successful conversion therapy, the general condition of the patient was good, and has been carefully followed for 4 months to date without any evidence of further recurrences. LESSONS This case showed that even if atezolizumab plus bevacizumab is not effective, multidisciplinary treatment such as LEN and conversion surgery is possible. Given the efficacy of LEN after atezolizumab plus bevacizumab, it is important to consider that there is a possibility of cure even when first-line treatment is not effective for a patient with unresectable HCC.
Collapse
Affiliation(s)
- Shigeki Yano
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Yusuke Johira
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Ryoichi Miura
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Masanari Kosaka
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Yuki Shirane
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Serami Murakami
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Kei Amioka
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Kensuke Naruto
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Yuwa Ando
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Yumi Kosaka
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Kenji Yamaoka
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Kenichiro Kodama
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Shinsuke Uchikawa
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Hatsue Fujino
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Atsushi Ohno
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Takashi Nakahara
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Eisuke Murakami
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Wataru Okamoto
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Masami Yamauchi
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| | - Keiichi Mori
- Anatomical Pathology, Hiroshima University, Japan
| | | | - Shintaro Kuroda
- Gastroenterological and Transplant Surgery, Hiroshima University, Japan
| | | | - Hideki Ohdan
- Gastroenterological and Transplant Surgery, Hiroshima University, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Hiroshima University, Japan
| |
Collapse
|