1
|
Shen H, Lynch EM, Jameson N, Decarreau J, Shi C, Chen Z, Sheffler W, Xu M, De Yoreo JJ, Zalatan JG, Kollman JM, Baker D. De novo design of inducibly assembling multi-component filaments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645066. [PMID: 40196579 PMCID: PMC11974854 DOI: 10.1101/2025.03.26.645066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The design of multi-component nanomaterials is an outstanding challenge. Here, we describe the computational design of protein filaments with two or three distinct structural components that assemble into micron-scale, well-ordered fibers when mixed. CryoEM structure determination of four fiber designs was close to the computational design models. Filament assembly can be initiated by mixing the components, and modulated by addition and/or phosphorylation of designed regulatory subunits. This work demonstrates that regulatable multi-component protein filament systems can now be designed, opening the door to a wide range of engineered materials.
Collapse
Affiliation(s)
- Hao Shen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Eric M. Lynch
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Noel Jameson
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Chenyang Shi
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Zibo Chen
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - William Sheffler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Miaomiao Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, Jiangsu, China
| | - James J De Yoreo
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, USA
| | - Jesse G. Zalatan
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Justin M. Kollman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Liu WB, Yang WG, Wu J, Chen BB, Du YF, Niu JB, Song J, Zhang SY. Discovery of novel thienopyridine indole derivatives as inhibitors of tubulin polymerization targeting the colchicine-binding site with potent anticancer activities. Eur J Med Chem 2025; 286:117314. [PMID: 39874633 DOI: 10.1016/j.ejmech.2025.117314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Based on the molecular hybridization strategy, novel thienopyridine indole derivatives were designed and synthesized as tubulin polymerization inhibitors, and the in vitro antiproliferative potency on MGC-803, KYSE450 and HCT-116 cells was evaluated. Among them, compound 20b showed a broad-spectrum antiproliferative activity against 11 cancer cell lines, with IC50 values below 4 nmol/L. Notably, it demonstrated exceptional efficacy against MGC-803 (IC50 = 1.61 nmol/L) and HGC-27 (IC50 = 1.82 nmol/L) cells. Further mechanism explorations suggested that compound 20b could inhibit tubulin polymerization (IC50 = 2.505 μmol/L) by acting on the colchicine binding site, thereby disrupting intracellular microtubule networks and interfering with cell mitosis. In addition, compound 20b effectively inhibited the colony formation and cell migration activities, and induced G2/M phase cycle arrest and apoptosis in MGC-803 and HGC-27 cells. Besides, compound 20b also displayed potent anti-angiogenesis effects on HUVECs. Importantly, compound 20b demonstrated oral efficacy in inhibiting tumor growth with a TGI of 45.8 % (5 mg/kg/qod) in the mouse xenograft model bearing MGC-803 cells, surpassing that of CA-4 (TGI of 27.1 % at 20 mg/kg/qod), as well as also exhibited a good safety profile. Therefore, these results suggested that the thienopyridine indole derivative 20b represents a novel tubulin inhibitor with potent anticancer efficacy that inhibits gastric cancers.
Collapse
Affiliation(s)
- Wen-Bo Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wei-Guang Yang
- Children's Hospital Affiliated of Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450000, China
| | - Ji Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Bing-Bing Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi-Fei Du
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Jin L, Qin Y, Zhao Y, Zhou X, Zeng Y. Endothelial cytoskeleton in mechanotransduction and vascular diseases. J Biomech 2025; 182:112579. [PMID: 39938443 DOI: 10.1016/j.jbiomech.2025.112579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The cytoskeleton is an important structural component that regulates various aspects of cell morphology, movement, and intracellular signaling. It plays a pivotal role in the cellular response to biomechanical stimuli, particularly in endothelial cells, which are critical for vascular homeostasis and the pathogenesis of cardiovascular diseases. Mechanical forces, such as shear and tension, activate intracellular signaling cascades that regulate transcription, translation, and cellular behaviors. Despite extensive research into cytoskeletal functions, the precise mechanisms by which the cytoskeleton transduces mechanical signals remain incompletely understood. This review focuses on the role of cytoskeletal components in membrane, cytoplasm, and nucleus in mechanotransduction, with an emphasis on their structure, mechanical and biological behaviors, dynamic interactions, and response to mechanical forces. The collaboration between membrane cytoskeleton, cytoplasmic cytoskeleton, and nucleoskeleton is indispensable for endothelial cells to respond to mechanical stimuli. Understanding their mechanoresponsive mechanisms is essential for advancing therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yunran Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
4
|
Chinnathambi S, Rangappa N, Chandrashekar M. Internalization of extracellular Tau oligomers in Alzheimer's disease. Adv Clin Chem 2025; 126:1-29. [PMID: 40185532 DOI: 10.1016/bs.acc.2025.01.005] [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: 04/07/2025]
Abstract
A key factor in the progression of Alzheimer's disease (AD) is internalization of extracellular Tau oligomers (ecTauOs) by neuroglial cells. Aberrant hyperphosphorylation of Tau results in their dissociation from microtubules and formation of toxic intracellular Tau oligomers (icTauOs). These are subsequently released to the extracellular space following neuronal dysfunction and death. Although receptor mediated internalization of these ecTauOs by other neurons, microglia and astrocytes can facilitate elimination, incomplete degradation thereof promotes inflammation, exacerbates pathologic spread and accelerates neurodegeneration. Targeting Tau oligomer degradation pathways, blocking internalization receptors, and mitigating neuroinflammation are proposed as therapeutic strategies to control Tau propagation and toxicity. This review highlights the urgent need for innovative approaches to prevent the spread of Tau pathology, emphasizing its implications for AD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India.
| | - Nagaraj Rangappa
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Madhura Chandrashekar
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| |
Collapse
|
5
|
Chew YM, Cross RA. Structural switching of tubulin in the microtubule lattice. Biochem Soc Trans 2025; 53:BST20240360. [PMID: 39910801 DOI: 10.1042/bst20240360] [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: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Microtubule (MT) dynamic instability, a cycle of growth, catastrophe, shrinkage and rescue, is driven by the switching of tubulin between two structural states, one stabilised by GTP and the other by GDP. Recent work has uncovered the ancient origins of this structural switch and revealed further fundamental elements of microtubule dynamic instability, whereby switching can be brought about by a range of allosteric effectors, propagate deep within the lattice of assembled MTs, and profoundly affect MT function. Here, we review evidence for structural switching within the MT lattice and discuss current ideas about its mechanisms.
Collapse
Affiliation(s)
- Yean-Ming Chew
- Centre for Mechanochemical Cell Biology, University of Warwick, Warwick Medical School, Coventry CV4 7LA, U.K
| | - Robert A Cross
- Centre for Mechanochemical Cell Biology, University of Warwick, Warwick Medical School, Coventry CV4 7LA, U.K
| |
Collapse
|
6
|
Montecinos F, Eren E, Watts NR, Sackett DL, Wingfield PT. Structure of blood cell-specific tubulin and demonstration of dimer spacing compaction in a single protofilament. J Biol Chem 2025; 301:108132. [PMID: 39725029 PMCID: PMC11791314 DOI: 10.1016/j.jbc.2024.108132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Microtubule (MT) function plasticity originates from its composition of α- and β-tubulin isotypes and the posttranslational modifications of both subunits. Aspects such as MT assembly dynamics, structure, and anticancer drug binding can be modulated by αβ-tubulin heterogeneity. However, the exact molecular mechanism regulating these aspects is only partially understood. A recent insight is the discovery of expansion and compaction of the MT lattice, which can occur via fine modulation of dimer longitudinal spacing mediated by GTP hydrolysis, taxol binding, protein binding, or isotype composition. Here, we report the first structure of the blood cell-specific α1/β1-tubulin isolated from the marginal band of chicken erythrocytes (ChET) determined to a resolution of 3.2 Å by cryo-EM. We show that ChET rings induced with cryptophycin-52 (Cp-52) are smaller in diameter than HeLa cell line tubulin (HeLaT) rings induced with Cp-52 and composed of the same number of heterodimers. We observe compacted interdimer and intradimer curved protofilament interfaces, characterized by shorter distances between ChET subunits and accompanied by conformational changes in the β-tubulin subunit. The compacted ChET interdimer interface brings more residues near the Cp-52 binding site. We measured the Cp-52 apparent binding affinities of ChET and HeLaT by mass photometry, observing small differences, and detected the intermediates of the ring assembly reaction. These findings demonstrate that compaction/expansion of dimer spacing can occur in a single protofilament context and that the subtle structural differences between tubulin isotypes can modulate tubulin small molecule binding.
Collapse
Affiliation(s)
- Felipe Montecinos
- Protein Expression Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elif Eren
- Protein Expression Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Norman R Watts
- Protein Expression Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Dan L Sackett
- Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA.
| | - Paul T Wingfield
- Protein Expression Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
7
|
Chen G, Qi H, Jiang L, Sun S, Zhang J, Yu J, Liu F, Zhang Y, Du S. Integrating single-cell RNA-Seq and machine learning to dissect tryptophan metabolism in ulcerative colitis. J Transl Med 2024; 22:1121. [PMID: 39707393 DOI: 10.1186/s12967-024-05934-w] [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: 07/30/2024] [Accepted: 12/01/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a persistent inflammatory bowels disease (IBD) characterized by immune response dysregulation and metabolic disruptions. Tryptophan metabolism has been believed as a significant factor in UC pathogenesis, with specific metabolites influencing immune modulation and gut microbiota interactions. However, the precise regulatory mechanisms and key genes involved remain unclear. METHODS AUCell, Ucell, and other functional enrichment algorithms were utilized to determine the activation patterns of tryptophan metabolism at the UC cell level. Differential analysis identified key genes associated with tryptophan metabolism. Five machine learning algorithms, including Random Forest, Boruta algorithm, LASSO, SVM-RFE, and GBM were integrated to identify and categorize disease-specific characteristic genes. RESULTS We observed significant heterogeneity in tryptophan metabolism activity across cell types in UC, with the highest activity levels in macrophages and fibroblasts. Among the key tryptophan metabolism-related genes, CTSS, S100A11, and TUBB were predominantly expressed in macrophages and significantly upregulated in UC, highlighting their involvement in immune dysregulation and inflammation. Cross-analysis with bulk RNA data confirmed the consistent upregulation of these genes in UC samples, highly indicating their relevance in UC pathology and potential as targets for therapeutic intervention. CONCLUSIONS This study is the first to reveal the heterogeneity of tryptophan metabolism at the single-cell level in UC, with macrophages emerging as key contributors to inflammatory processes. The identification of CTSS, S100A11, and TUBB as key regulators of tryptophan metabolism in UC underscores their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Guorong Chen
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongying Qi
- Department of Spleen and Stomach Diseases of Traditional Chinese Medicine, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Li Jiang
- Department of Endocrinology, Aviation General Hospital, Beijing, 100025, China
| | - Shijie Sun
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Junhai Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Jiali Yu
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Fang Liu
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Yanli Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China.
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China.
| |
Collapse
|
8
|
Sun Z, Wang X, Peng C, Dai L, Wang T, Zhang Y. Regulation of cytoskeleton dynamics and its interplay with force in plant cells. BIOPHYSICS REVIEWS 2024; 5:041307. [PMID: 39606182 PMCID: PMC11596143 DOI: 10.1063/5.0201899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
The plant cytoskeleton is an intricate network composed of actin filaments and microtubules. The cytoskeleton undergoes continuous dynamic changes that provide the basis for rapidly responding to intrinsic and extrinsic stimuli, including mechanical stress. Microtubules can respond to alterations of mechanical stress and reorient along the direction of maximal tensile stress in plant cells. The cytoskeleton can also generate driving force for cytoplasmic streaming, organelle movement, and vesicle transportation. In this review, we discuss the progress of how the plant cytoskeleton responds to mechanical stress. We also summarize the roles of the cytoskeleton in generating force that drive organelles and nuclear transportation in plant cells. Finally, some hypotheses concerning the link between the roles of the cytoskeleton in force response and organelle movement, as well as several key questions that remain to be addressed in the field, are highlighted.
Collapse
Affiliation(s)
- Zhenping Sun
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Xueqing Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Chaoyong Peng
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | | | - Ting Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yi Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
9
|
Ammar Khodja L, Campanacci V, Lippens G, Gigant B. The structure of a Tau fragment bound to tubulin prompts new hypotheses on Tau mechanism and oligomerization. PNAS NEXUS 2024; 3:pgae487. [PMID: 39534653 PMCID: PMC11554759 DOI: 10.1093/pnasnexus/pgae487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Tau is a protein involved in the regulation of axonal microtubules in neurons. In pathological conditions, it forms filamentous aggregates which are molecular markers of neurodegenerative diseases known as tauopathies. Structures of Tau in fibrils or bound to the microtubule have been reported. We present here a structure of a Tau construct comprising the PHF6 motif, an oligopeptide involved in Tau aggregation, as a complex with tubulin. This Tau fragment binds as a dimer to a new site which, when transposed to the microtubule, would correspond to a pore between protofilaments. These results raise new hypotheses on Tau-induced microtubule assembly and stabilization and on Tau oligomerization.
Collapse
Affiliation(s)
- Liza Ammar Khodja
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Valérie Campanacci
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Guy Lippens
- CNRS, TBI, Université de Toulouse, INRAE, INSA, 31077 Toulouse, France
| | - Benoît Gigant
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| |
Collapse
|
10
|
Ali F, Zhao Y, Ali A, Waseem M, Arif MAR, Shah OU, Liao L, Wang Z. Omics-Driven Strategies for Developing Saline-Smart Lentils: A Comprehensive Review. Int J Mol Sci 2024; 25:11360. [PMID: 39518913 PMCID: PMC11546581 DOI: 10.3390/ijms252111360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
A number of consequences of climate change, notably salinity, put global food security at risk by impacting the development and production of lentils. Salinity-induced stress alters lentil genetics, resulting in severe developmental issues and eventual phenotypic damage. Lentils have evolved sophisticated signaling networks to combat salinity stress. Lentil genomics and transcriptomics have discovered key genes and pathways that play an important role in mitigating salinity stress. The development of saline-smart cultivars can be further revolutionized by implementing proteomics, metabolomics, miRNAomics, epigenomics, phenomics, ionomics, machine learning, and speed breeding approaches. All these cutting-edge approaches represent a viable path toward creating saline-tolerant lentil cultivars that can withstand climate change and meet the growing demand for high-quality food worldwide. The review emphasizes the gaps that must be filled for future food security in a changing climate while also highlighting the significant discoveries and insights made possible by omics and other state-of-the-art biotechnological techniques.
Collapse
Affiliation(s)
- Fawad Ali
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, China; (F.A.); (Y.Z.); (M.W.); (O.U.S.)
| | - Yiren Zhao
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, China; (F.A.); (Y.Z.); (M.W.); (O.U.S.)
| | - Arif Ali
- Department of Plant Sciences, Quaid-I-Azam University, Islamabad 45320, Pakistan;
| | - Muhammad Waseem
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, China; (F.A.); (Y.Z.); (M.W.); (O.U.S.)
| | - Mian A. R. Arif
- Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C, PIEAS), Jhang Road, Faisalabad 38000, Pakistan;
| | - Obaid Ullah Shah
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, China; (F.A.); (Y.Z.); (M.W.); (O.U.S.)
| | - Li Liao
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, China; (F.A.); (Y.Z.); (M.W.); (O.U.S.)
| | - Zhiyong Wang
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, China; (F.A.); (Y.Z.); (M.W.); (O.U.S.)
| |
Collapse
|
11
|
Cheng L, Peng JZ, Li SW, Bie ZX, Li XG. Acute-phase plasma proteomics of rabbit lung VX2 tumors treated by image-guided microwave ablation. Front Oncol 2024; 14:1435256. [PMID: 39252952 PMCID: PMC11381224 DOI: 10.3389/fonc.2024.1435256] [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: 05/20/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024] Open
Abstract
Purpose To explore the plasma proteomic changes of rabbit lung VX2 tumors treated by microwave ablation, and to explore the molecular pathway mechanisms that may be involved. Methods New Zealand white rabbits were inoculated with VX2 tumor cell suspension in the right lower lung and treated with microwave ablation after 2-3 weeks of tumor formation. Blood was collected at 5 time points (TP1~TP5) before and after ablation by cardiac blood sampling and pre-treated before proteomic analysis. The plasma proteome was analyzed by Data-Independent Acquisition (DIA). Results Different molecular pathways were activated at different time points:(i) TP1vsTP2: more proteins were down-regulated and enrichment analysis showed that the proteasome pathway was activated. The abnormal protein folding process involved in this pathway is closely related to the process of tumor development. (ii) TP2vsTP3: more proteins were up-regulated although the number of differentially differentiated proteins was lower and enrichment analysis showed that the phagosome pathway was activated. After microwave ablation inactivates tumor cells, it activates the phagosomal pathway for immune clearance of necrotic tumor tissue. (iii) TP3vsTP4: more down-regulated proteins, enrichment analysis showed that cysteine and methionine metabolism pathway was activated. Decreased metabolism of these amino acids suggests that cancer progression may be blocked after microwave ablation therapy. (iv) TP4vsTP5: the number of differential proteins was less and more down-regulated proteins, enrichment analysis showed that glutathione metabolism and metabolism of xenobiotics by cytochrome P450 pathway were activated. The down-regulated proteins in this pathway may suggest that microwave ablation may have reduced resistance to certain chemotherapeutic agents following. Conclusions In the process of lung cancer treatment by microwave ablation, the changes of proteins on the possible molecular pathways at each time point are related to lung cancer, and not only involve some simple inflammatory reactions, and some of the proteins released by destroying the tumor cells can be used as possible drug binding sites and reduce drug resistance.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jin-Zhao Peng
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng-Wei Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Xin Bie
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Guang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Adelman JW, Sukowaty AT, Partridge KJ, Gawrys JE, Terhune SS, Ebert AD. Stabilizing microtubules aids neurite structure and disrupts syncytia formation in human cytomegalovirus-infected human forebrain neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608340. [PMID: 39229072 PMCID: PMC11370344 DOI: 10.1101/2024.08.16.608340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Human cytomegalovirus (HCMV) is a prolific human herpesvirus that infects most individuals by adulthood. While typically asymptomatic in adults, congenital infection can induce serious neurological symptoms including hearing loss, visual deficits, cognitive impairment, and microcephaly in 10-15% of cases. HCMV has been shown to infect most neural cells with our group recently demonstrating this capacity in stem cell-derived forebrain neurons. Infection of neurons induces deleterious effects on calcium dynamics and electrophysiological function paired with gross restructuring of neuronal morphology. Here, we utilize an iPSC-derived model of the human forebrain to demonstrate how HCMV infection induces syncytia, drives neurite retraction, and remodels microtubule networks to promote viral production and release. We establish that HCMV downregulates microtubule associated proteins at 14 days postinfection while simultaneously sparing other cytoskeletal elements, and this includes HCMV-driven alterations to microtubule stability. Further, we pharmacologically modulate microtubule dynamics using paclitaxel (stabilize) and colchicine (destabilize) to examine the effects on neurite structure, syncytial morphology, assembly compartment formation, and viral release. With paclitaxel, we found improvement of neurite outgrowth with a corresponding disruption to HCMV-induced syncytia formation and Golgi network disruptions but with limited impact on viral titers. Together, these data suggest that HCMV infection-induced disruption of microtubules in human cortical neurons can be partially mitigated with microtubule stabilization, suggesting a potential avenue for future neuroprotective therapeutic exploration.
Collapse
Affiliation(s)
- Jacob W Adelman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andrew T Sukowaty
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kaitlyn J Partridge
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jessica E. Gawrys
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott S. Terhune
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Marquette University and Medical College of Wisconsin Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
13
|
Yang H, Deng Y, Dong Y, Ma Y, Yang L. Identification and Validation of Prognostic Markers for Endometriosis-Associated Ovarian Cancer. Int J Med Sci 2024; 21:1903-1914. [PMID: 39113897 PMCID: PMC11302560 DOI: 10.7150/ijms.97024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Background: Growing evidence suggests that endometriosis (EMs) is a risk factor for endometriosis-associated ovarian cancer (EAOC). The aim was to identify and validate gene signatures associated with EMs that may serve as potential biomarkers for evaluating the prognosis of patients with EAOC. Methods: The data of EMs and control samples was obtained from GEO database. The weighted gene co-expression network analysis (WGCNA) identified modular genes significantly associated with EMs. The KEGG pathway and GO functional enrichment analyses were also performed. Univariate Cox regression analysis was conducted to screen marker genes associated with the prognosis of EAOC patients. Finally, RT-qPCR and immunohistochemical verified the expression of ADAMTS19 and TUBB in normal ovarian and EAOC tissues, and the biological functions of ADAMTS19 and TUBB were preliminarily explored by CCK8 and Transwell assays. Results: The WGCNA identified 2 co-expression modules, which in total included 615 genes, and 7642 differentially expressed genes (DEGs) were detected thorough analysis of the EAOC dataset. After taking the intersection of 615 modular genes and 7642 DEGs, 214 shared genes were obtained, and univariate COX regression analysis pointed 10 genes associated with the prognosis of EAOC. Moreover, it was demonstrated by RT-qPCR and immunohistochemical staining experiments that ADAMTS19 expression was elevated, while TUBB expression was reduced in EAOC compared with normal ovarian cells and tissues. Finally, cell experiments revealed that ADAMTS19 promoted the proliferation and invasion in EAOC cells, while overexpression of TUBB inhibited these processes. Conclusions: The present study identified and validated new EMs-associated gene markers, which could serve as potential biomarkers for assessing the prognostic risk of EAOC patients. In addition, some of these genes may have significance as novel therapeutic targets and could be used to guide clinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Lihua Yang
- Department of Gynecology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China
| |
Collapse
|
14
|
Yon WJ, Ha T, Zheng Y, Pedersen RTA. A tubulin-binding protein that preferentially binds to GDP-tubulin and promotes GTP exchange. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.09.539990. [PMID: 37214866 PMCID: PMC10197657 DOI: 10.1101/2023.05.09.539990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
α- and β-tubulin form heterodimers, with GTPase activity, that assemble into microtubules. Like other GTPases, the nucleotide-bound state of tubulin heterodimers controls whether the molecules are in a biologically active or inactive state. While α-tubulin in the heterodimer is constitutively bound to GTP, β-tubulin can be bound to either GDP (GDP-tubulin) or GTP (GTP-tubulin). GTP-tubulin hydrolyzes its GTP to GDP following assembly into a microtubule and, upon disassembly, must exchange its bound GDP for GTP to participate in subsequent microtubule polymerization. Tubulin dimers have been shown to exhibit rapid intrinsic nucleotide exchange in vitro, leading to a commonly accepted belief that a tubulin guanine nucleotide exchange factor (GEF) may be unnecessary in cells. Here, we use quantitative binding assays to show that BuGZ, a spindle assembly factor, binds tightly to GDP-tubulin, less tightly to GTP-tubulin, and weakly to microtubules. We further show that BuGZ promotes the incorporation of GTP into tubulin using a nucleotide exchange assay. The discovery of a tubulin GEF suggests a mechanism that may aid rapid microtubule assembly dynamics in cells.
Collapse
Affiliation(s)
- Wesley J Yon
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
- Cell, Molecular, Developmental Biology, and Biophysics Program, Johns Hopkins University, Baltimore, MD, USA
| | - Taekjip Ha
- Cell, Molecular, Developmental Biology, and Biophysics Program, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
- Cell, Molecular, Developmental Biology, and Biophysics Program, Johns Hopkins University, Baltimore, MD, USA
| | - Ross T A Pedersen
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| |
Collapse
|
15
|
Abdelmegeed H, Abdel Ghany LMA, Youssef A, El-Etrawy AAS, Ryad N. Exploring the antitumor potential of novel quinoline derivatives via tubulin polymerization inhibition in breast cancer; design, synthesis and molecular docking. RSC Adv 2024; 14:22092-22112. [PMID: 39005243 PMCID: PMC11240139 DOI: 10.1039/d4ra04371e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
A series of quinoline derivatives was designed and synthesized as novel tubulin inhibitors targeting the colchicine binding site. All the rationalized compounds 3a-e, 4a-e, 5a-e, and 6a-e have been chosen for screening their cytotoxic activity against 60 cell lines by NCI. Compounds 3b, 3c, 4c, 5c and 6c demonstrated the most notable antitumor activity against almost all cell lines. Compound 4c emerged as the most potent compound as an antiproliferative agent. This compound was subsequently chosen for five-dose testing and it exhibited remarkable broad-spectrum efficacy with strong antitumor activity against several cell lines. Compound 4c significantly induced cell cycle arrest in MDA-MB-231 cells at G2 and M phases where the cell population increased dramatically to 22.84% compared to the untreated cells at 10.42%. It also increased the population in MDA-MB-231 cells at both early and late stages of apoptosis. Compound 4c can successfully inhibit tubulin polymerization with an IC50 value of 17 ± 0.3 μM. The β-tubulin mRNA levels were notably reduced in MDA-MB-231 cells treated with compound 4c which is similar to the effect observed with colchicine treatment. Docking studies revealed that compound 4c interacted well with crucial amino acids in the active site.
Collapse
Affiliation(s)
- Heba Abdelmegeed
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Giza 12622 Egypt
| | - Lina M A Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City, P.O. Box 77 Giza Egypt
| | - Amira Youssef
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City, P.O. Box 77 Giza Egypt
| | - Abd-Allah S El-Etrawy
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City, P.O. Box 77 Giza Egypt
- Department of Chemistry, Basic Science, Misr University for Science and Technology (MUST) 6th of October City, P.O. Box 77 Giza Egypt
| | - Noha Ryad
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City, P.O. Box 77 Giza Egypt
| |
Collapse
|
16
|
Abel AC, Mühlethaler T, Dessin C, Schachtsiek T, Sammet B, Sharpe T, Steinmetz MO, Sewald N, Prota AE. Bridging the maytansine and vinca sites: Cryptophycins target β-tubulin's T5-loop. J Biol Chem 2024; 300:107363. [PMID: 38735475 PMCID: PMC11190709 DOI: 10.1016/j.jbc.2024.107363] [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: 01/16/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024] Open
Abstract
Cryptophycins are microtubule-targeting agents (MTAs) that belong to the most potent antimitotic compounds known to date; however, their exact molecular mechanism of action remains unclear. Here, we present the 2.2 Å resolution X-ray crystal structure of a potent cryptophycin derivative bound to the αβ-tubulin heterodimer. The structure addresses conformational issues present in a previous 3.3 Å resolution cryo-electron microscopy structure of cryptophycin-52 bound to the maytansine site of β-tubulin. It further provides atomic details on interactions of cryptophycins, which had not been described previously, including ones that are in line with structure-activity relationship studies. Interestingly, we discovered a second cryptophycin-binding site that involves the T5-loop of β-tubulin, a critical secondary structure element involved in the exchange of the guanosine nucleotide and in the formation of longitudinal tubulin contacts in microtubules. Cryptophycins are the first natural ligands found to bind to this new "βT5-loop site" that bridges the maytansine and vinca sites. Our results offer unique avenues to rationally design novel MTAs with the capacity to modulate T5-loop dynamics and to simultaneously engage multiple β-tubulin binding sites.
Collapse
Affiliation(s)
- Anne-Catherine Abel
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland; Biozentrum, University of Basel, Basel, Switzerland
| | | | - Cedric Dessin
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Thomas Schachtsiek
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Benedikt Sammet
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | | | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland; Biozentrum, University of Basel, Basel, Switzerland
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany.
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland.
| |
Collapse
|
17
|
Chakraborty J, Poddar S, Dutta S, Bahulekar V, Harne S, Srinivasan R, Gayathri P. Dynamics of interdomain rotation facilitates FtsZ filament assembly. J Biol Chem 2024; 300:107336. [PMID: 38718863 PMCID: PMC11157280 DOI: 10.1016/j.jbc.2024.107336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024] Open
Abstract
FtsZ, the tubulin homolog essential for bacterial cell division, assembles as the Z-ring at the division site, and directs peptidoglycan synthesis by treadmilling. It is unclear how FtsZ achieves kinetic polarity that drives treadmilling. To obtain insights into fundamental features of FtsZ assembly dynamics independent of peptidoglycan synthesis, we carried out structural and biochemical characterization of FtsZ from the cell wall-less bacteria, Spiroplasma melliferum (SmFtsZ). Interestingly the structures of SmFtsZ, bound to GDP and GMPPNP respectively, were captured as domain swapped dimers. SmFtsZ was found to be a slower GTPase with a higher critical concentration (CC) compared to Escherichia coli FtsZ (EcFtsZ). In FtsZs, a conformational switch from R-state (close) to T-state (open) favors polymerization. We identified that Phe224, located at the interdomain cleft of SmFtsZ, is crucial for R- to T-state transition. SmFtsZF224M exhibited higher GTPase activity and lower CC, whereas the corresponding EcFtsZM225F resulted in cell division defects in E. coli. Our results demonstrate that relative rotation of the domains is a rate-limiting step of polymerization. Our structural analysis suggests that the rotation is plausibly triggered upon addition of a GTP-bound monomer to the filament through interaction of the preformed N-terminal domain (NTD). Hence, addition of monomers to the NTD-exposed end of filament is slower in comparison to the C-terminal domain (CTD) end, thus explaining kinetic polarity. In summary, the study highlights the importance of interdomain interactions and conformational changes in regulating FtsZ assembly dynamics.
Collapse
Affiliation(s)
- Joyeeta Chakraborty
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Sakshi Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| | - Soumyajit Dutta
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Vaishnavi Bahulekar
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Shrikant Harne
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| | - Pananghat Gayathri
- Biology Division, Indian Institute of Science Education and Research, Pune, India.
| |
Collapse
|
18
|
Liu Z, Yang Z, Ablise M. Design and synthesis of novel imidazole-chalcone derivatives as microtubule protein polymerization inhibitors to treat cervical cancer and reverse cisplatin resistance. Bioorg Chem 2024; 147:107310. [PMID: 38583249 DOI: 10.1016/j.bioorg.2024.107310] [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: 01/19/2024] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024]
Abstract
Using the licochalcone moiety as a lead compound scaffold, 16 novel imidazole-chalcone derivatives were designed and synthesized as microtubule protein polymerization inhibitors. The proliferation inhibitory activities of the derivatives against SiHa (human cervical squamous cell carcinoma), C-33A (human cervical cancer), HeLa (human cervical cancer), HeLa/DDP (cisplatin-resistant human cervical cancer), and H8 (human cervical epithelial immortalized) cells were evaluated. Compound 5a exhibited significant anticancer activity with IC50 values ranging from 2.28 to 7.77 μM and a resistance index (RI) of 1.63, while showing minimal toxicity to normal H8 cells. When compound 5a was coadministered with cisplatin, the RI of cisplatin to HeLa/DDP cells decreased from 6.04 to 2.01, while compound 5a enhanced the fluorescence intensity of rhodamine 123 in HeLa/DDP cells. Further studies demonstrated that compound 5a arrested cells at the G2/M phase, induced apoptosis, reduced colony formation, inhibited cell migration, and inhibited cell invasion. Preliminary mechanistic studies revealed that compound 5a decreased the immunofluorescence intensity of α-/β-tubulin in cancer cells, reduced the expression of polymerized α-/β-tubulin, and increased the expression of depolymerized α-/β-tubulin. Additionally, the molecular docking results demonstrate that compound 5a can interact with the tubulin colchicine binding site and generate multiple types of interactions. These results suggested that compound 5a has anticancer effects and significantly reverses cervical cancer resistance to cisplatin, which may be related to its inhibition of microtubule and P-glycoprotein (P-gp) activity.
Collapse
Affiliation(s)
- Zhengye Liu
- The Xinjiang Key Laboratory of Natural Medicine Active Components and Drug Release Technology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Zheng Yang
- The Xinjiang Key Laboratory of Natural Medicine Active Components and Drug Release Technology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Mourboul Ablise
- The Xinjiang Key Laboratory of Natural Medicine Active Components and Drug Release Technology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
19
|
Yancheva D, Argirova M, Georgieva I, Milanova V, Guncheva M, Rangelov M, Todorova N, Tzoneva R. Antiproliferative and Pro-Apoptotic Activity and Tubulin Dynamics Modulation of 1 H-Benzimidazol-2-yl Hydrazones in Human Breast Cancer Cell Line MDA-MB-231. Molecules 2024; 29:2400. [PMID: 38792260 PMCID: PMC11123699 DOI: 10.3390/molecules29102400] [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: 03/16/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: The aim of the work is the evaluation of in vitro antiproliferative and pro-apoptotic activity of four benzimidazole derivatives containing colchicine-like and catechol-like moieties with methyl group substitution in the benzimidazole ring against highly invasive breast cancer cell line MDA-MB-231 and their related impairment of tubulin dynamics. (2) Methods: The antiproliferative activity was assessed with the MTT assay. Alterations in tubulin polymerization were evaluated with an in vitro tubulin polymerization assay and a docking analysis. (3) Results: All derivatives showed time-dependent cytotoxicity with IC50 varying from 40 to 60 μM after 48 h and between 13 and 20 μM after 72 h. Immunofluorescent and DAPI staining revealed the pro-apoptotic potential of benzimidazole derivatives and their effect on tubulin dynamics in living cells. Compound 5d prevented tubulin aggregation and blocked mitosis, highlighting the importance of the methyl group and the colchicine-like fragment. (4) Conclusions: The benzimidazole derivatives demonstrated moderate cytotoxicity towards MDA-MB-231 by retarding the initial phase of tubulin polymerization. The derivative 5d containing a colchicine-like moiety and methyl group substitution in the benzimidazole ring showed potential as an antiproliferative agent and microtubule destabilizer by facilitating faster microtubule aggregation and disrupting cellular and nuclear integrity.
Collapse
Affiliation(s)
- Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 9, 1113 Sofia, Bulgaria; (M.A.); (M.G.); (M.R.)
| | - Maria Argirova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 9, 1113 Sofia, Bulgaria; (M.A.); (M.G.); (M.R.)
| | - Irina Georgieva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 21, 1113 Sofia, Bulgaria; (I.G.); (V.M.)
| | - Vanya Milanova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 21, 1113 Sofia, Bulgaria; (I.G.); (V.M.)
| | - Maya Guncheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 9, 1113 Sofia, Bulgaria; (M.A.); (M.G.); (M.R.)
| | - Miroslav Rangelov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 9, 1113 Sofia, Bulgaria; (M.A.); (M.G.); (M.R.)
| | - Nadezhda Todorova
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 2 Gagarin Str., 1113 Sofia, Bulgaria;
| | - Rumiana Tzoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Build. 21, 1113 Sofia, Bulgaria; (I.G.); (V.M.)
| |
Collapse
|
20
|
Zhang L, Fu Y, Tong Y, Xie G, Deng S. Dynamic DNA Assembly by Programmable Hybridization Chain Reaction Mimicking Tubulin. NANO LETTERS 2024; 24:2603-2610. [PMID: 38349971 DOI: 10.1021/acs.nanolett.3c04852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Dynamic biological structures involve the continual turnover of molecules within supramolecular assemblies such as tubulin. Inspired by dynamic biology self-organizing systems, we build an artificial dynamic structure based on DNA nanotechnology through a nonequilibrium chemical system. Herein, a metastable domain (MD), essentially a stem-loop structure, was introduced into DNA hairpins within hybridization chain reaction (HCR), thereby imparting dynamic activity to the DNA polymers. Hairpins with MD thermodynamically assemble to a high-energy polymer in the presence of trigger strands. The polymer can relax back to the stable unassembled state once the invader is added and finally relax to the activated hairpin by an anti-invader. Reversible assembly/disassembly of the HCR is achieved through invader/anti-invader cycles. We accomplished kinetic modulation, reversible conformational switching, cascading regulation, and enzyme activity control through the MD-HCR. We believe that the design of the MD-HCR could inspire the development of autonomous biological functions within artificial systems.
Collapse
Affiliation(s)
- Li Zhang
- Department of Forensic Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yao Fu
- Department of Forensic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yanqiu Tong
- School of Tourism and Media, Chongqing Jiaotong University, Chongqing 400016, China
| | - Guoming Xie
- Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shixiong Deng
- Department of Forensic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
21
|
Sati P, Sharma E, Dhyani P, Attri DC, Rana R, Kiyekbayeva L, Büsselberg D, Samuel SM, Sharifi-Rad J. Paclitaxel and its semi-synthetic derivatives: comprehensive insights into chemical structure, mechanisms of action, and anticancer properties. Eur J Med Res 2024; 29:90. [PMID: 38291541 PMCID: PMC10826257 DOI: 10.1186/s40001-024-01657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Cancer is a disease that can cause abnormal cell growth and can spread throughout the body. It is among the most significant causes of death worldwide, resulting in approx. 10 million deaths annually. Many synthetic anticancer drugs are available, but they often come with side effects and can interact negatively with other medications. Additionally, many chemotherapy drugs used for cancer treatment can develop resistance and harm normal cells, leading to dose-limiting side effects. As a result, finding effective cancer treatments and developing new drugs remains a significant challenge. However, plants are a potent source of natural products with the potential for cancer treatment. These biologically active compounds may be the basis for enhanced or less toxic derivatives. Herbal medicines/phytomedicines, or plant-based drugs, are becoming more popular in treating complicated diseases like cancer due to their effectiveness and are a particularly attractive option due to their affordability, availability, and lack of serious side effects. They have broad applicability and therapeutic efficacy, which has spurred scientific research into their potential as anticancer agents. This review focuses on Paclitaxel (PTX), a plant-based drug derived from Taxus sp., and its ability to treat specific tumors. PTX and its derivatives are effective against various cancer cell lines. Researchers can use this detailed information to develop effective and affordable treatments for cancer.
Collapse
Affiliation(s)
- Priyanka Sati
- Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Eshita Sharma
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Praveen Dhyani
- Institute for Integrated Natural Sciences, University of Koblenz, Koblenz, Germany
| | - Dharam Chand Attri
- Department of Botany, Central University of Jammu, Rahya-Suchani (Bagla), Jammu and Kashmir, India
| | - Rohit Rana
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Lashyn Kiyekbayeva
- Department of Pharmaceutical Technology, Pharmaceutical School, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar.
| | | |
Collapse
|
22
|
Jiang M, Zhou C, Wang S, Liu L, Zhang S, Wang L, Pan X. Identification of a Tetrahymena species infecting guppies, pathology, and expression of beta-tubulin during infection. Parasitol Res 2024; 123:104. [PMID: 38240890 DOI: 10.1007/s00436-024-08117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024]
Abstract
Tetrahymenosis is caused by the ciliated protozoan Tetrahymena and is responsible for serious economic losses to the aquaculture industry worldwide. However, information regarding the molecular mechanism leading to tetrahymenosis is limited. In previous transcriptome sequencing work, it was found that one of the two β-tubulin genes in T. pyriformis was significantly expressed in infected fish, we speculated that β-tubulin is involved in T. pyriformis infecting fish. Herein, the potential biological function of the β-tubulin gene in Tetrahymena species when establishing infection in guppies was investigated by cloning the full-length cDNA of this T. pyriformis β-tubulin (BTU1) gene. The full-length cDNA of T. pyriformis BTU1 gene was 1873 bp, and the ORF occupied 1134 bp, whereas 5' UTR 434 bp, and 3' UTR 305 bp whose poly (A) tail contained 12 bases. The predicted protein encoded by T. pyriformis BTU1 gene had a calculated molecular weight of 42.26 kDa and pI of 4.48. Moreover, secondary structure analysis and tertiary structure prediction of BTU1 protein were also conducted. In addition, morphology, infraciliature, phylogeny, and histopathology of T. pyriformis isolated from guppies from a fish market in Harbin were also investigated. Furthermore, qRT-PCR analysis and experimental infection assays indicated that the expression of BTU1 gene resulted in efficient cell proliferation during infection. Collectively, our data revealed that BTU1 is a key gene involved in T. pyriformis infection in guppies, and the findings discussed herein provide valuable insights for future studies on tetrahymenosis.
Collapse
Affiliation(s)
- Mingyue Jiang
- Laboratory of Protozoology, Harbin Normal University, Harbin, 150025, China
| | - Chunyu Zhou
- Laboratory of Protozoology, Harbin Normal University, Harbin, 150025, China
| | - Sihan Wang
- Laboratory of Protozoology, Harbin Normal University, Harbin, 150025, China
| | - Lihui Liu
- Laboratory of Protozoology, Harbin Normal University, Harbin, 150025, China
| | - Shuixian Zhang
- Panzhou No.4 Primary School, Panzhou, 553599, Guizhou Province, China
| | - Li Wang
- Laboratory of Protozoology, Harbin Normal University, Harbin, 150025, China
| | - Xuming Pan
- Laboratory of Protozoology, Harbin Normal University, Harbin, 150025, China.
| |
Collapse
|
23
|
Nammalwar B, Bunce RA. Recent Advances in Pyrimidine-Based Drugs. Pharmaceuticals (Basel) 2024; 17:104. [PMID: 38256937 PMCID: PMC10820437 DOI: 10.3390/ph17010104] [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: 11/27/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
Pyrimidines have become an increasingly important core structure in many drug molecules over the past 60 years. This article surveys recent areas in which pyrimidines have had a major impact in drug discovery therapeutics, including anti-infectives, anticancer, immunology, immuno-oncology, neurological disorders, chronic pain, and diabetes mellitus. The article presents the synthesis of the medicinal agents and highlights the role of the biological target with respect to the disease model. Additionally, the biological potency, ADME properties and pharmacokinetics/pharmacodynamics (if available) are discussed. This survey attempts to demonstrate the versatility of pyrimidine-based drugs, not only for their potency and affinity but also for the improved medicinal chemistry properties of pyrimidine as a bioisostere for phenyl and other aromatic π systems. It is hoped that this article will provide insight to researchers considering the pyrimidine scaffold as a chemotype in future drug candidates in order to counteract medical conditions previously deemed untreatable.
Collapse
Affiliation(s)
- Baskar Nammalwar
- Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA 92121, USA;
| | - Richard A. Bunce
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
24
|
Syed RA, Hayat M, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Kaleem I, Bashir S. Aging-Related Protein Alterations in the Brain. J Alzheimers Dis 2024; 99:S5-S22. [PMID: 38339930 DOI: 10.3233/jad-230801] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Aging is an intrinsic aspect of an organism's life cycle and is characterized by progressive physiological decline and increased susceptibility to mortality. Many age-associated disorders, including neurological disorders, are most commonly linked with the aging process, such as Alzheimer's disease (AD). This review aims to provide a comprehensive overview of the effects of aging and AD on the molecular pathways and levels of different proteins in the brain, including metalloproteins, neurotrophic factors, amyloid proteins, and tau proteins. AD is caused by the aggregation of amyloid proteins in the brain. Factors such as metal ions, protein ligands, and the oligomerization state of amyloid precursor protein significantly influence the proteolytic processing of amyloid-β protein precursor (AβPP). Tau, a disordered cytosolic protein, serves as the principal microtubule-associated protein in mature neurons. AD patients exhibit decreased levels of nerve growth factor within their nervous systems and cerebrospinal fluid. Furthermore, a significant increase in brain-derived neurotrophic factor resulting from the neuroprotective effect of glial cell line-derived neurotrophic factor suggests that the synergistic action of these proteins plays a role in inhibiting neuronal degeneration and atrophy. The mechanism through which Aβ and AβPP govern Cu2+ transport and their influence on Cu2+ and other metal ion pools requires elucidation in future studies. A comprehensive understanding of the influence of aging and AD on molecular pathways and varying protein levels may hold the potential for the development of novel diagnostic and therapeutic methods for the treatment of AD.
Collapse
Affiliation(s)
- Rafay Ali Syed
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Mahnoor Hayat
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Imdad Kaleem
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
25
|
Cui YJ, Zhou Y, Zhang XW, Dou BK, Ma CC, Zhang J. The discovery of water-soluble indazole derivatives as potent microtubule polymerization inhibitors. Eur J Med Chem 2023; 262:115870. [PMID: 37890199 DOI: 10.1016/j.ejmech.2023.115870] [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: 08/23/2023] [Revised: 09/26/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
Taking a previously discovered indazole derivative 1 as a lead, systematic structural modifications were performed with an indazole core at the 1- and 6-positions to improve its aqueous solubility. Among the designed indazole derivatives, 6-methylpyridin-3-yl indazole derivative 8l and 1H-indol-4-yl indazole derivative 8m exhibited high potency in the low nanomolar range against A549, Huh-7, and T24 cancer cells, including Taxol-resistant variant cells (A549/Tax). As a hydrochloride salt, 8l exhibited much improved aqueous solubility, and its log P value fell into a favorable range. In mechanistic studies, 8l impeded tubulin polymerization through interacting with the colchicine site, resulting in cell cycle arrest and cellular apoptosis. In addition, compared to lead compound 1, 8l reduced cell migration and led to more potent inhibition of tumor growth in vivo without apparent toxicity. In summary, indazole derivative 8l could work as a potential anticancer agent and deserves further investigation for cancer therapy.
Collapse
Affiliation(s)
- Ying-Jie Cui
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Yi Zhou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Xi-Wu Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Bao-Kai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Chen-Chen Ma
- Central Laboratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250012, China.
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China.
| |
Collapse
|
26
|
Abbaali I, Truong D, Day SD, Mushayeed F, Ganesh B, Haro-Ramirez N, Isles J, Nag H, Pham C, Shah P, Tomar I, Manel-Romero C, Morrissette NS. The tubulin database: Linking mutations, modifications, ligands and local interactions. PLoS One 2023; 18:e0295279. [PMID: 38064432 PMCID: PMC10707541 DOI: 10.1371/journal.pone.0295279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Microtubules are polymeric filaments, constructed of α-β tubulin heterodimers that underlie critical subcellular structures in eukaryotic organisms. Four homologous proteins (γ-, δ-, ε- and ζ-tubulin) additionally contribute to specialized microtubule functions. Although there is an immense volume of publicly available data pertaining to tubulins, it is difficult to assimilate all potentially relevant information across diverse organisms, isotypes, and categories of data. We previously assembled an extensive web-based catalogue of published missense mutations to tubulins with >1,500 entries that each document a specific substitution to a discrete tubulin, the species where the mutation was described and the associated phenotype with hyperlinks to the amino acid sequence and citation(s) for research. This report describes a significant update and expansion of our online resource (TubulinDB.bio.uci.edu) to nearly 18,000 entries. It now encompasses a cross-referenced catalog of post-translational modifications (PTMs) to tubulin drawn from public datasets, primary literature, and predictive algorithms. In addition, tubulin protein structures were used to define local interactions with bound ligands (GTP, GDP and diverse microtubule-targeting agents) and amino acids at the intradimer interface, within the microtubule lattice and with associated proteins. To effectively cross-reference these datasets, we established a universal tubulin numbering system to map entries into a common framework that accommodates specific insertions and deletions to tubulins. Indexing and cross-referencing permitted us to discern previously unappreciated patterns. We describe previously unlinked observations of loss of PTM sites in the context of cancer cells and tubulinopathies. Similarly, we expanded the set of clinical substitutions that may compromise MAP or microtubule-motor interactions by collecting tubulin missense mutations that alter amino acids at the interface with dynein and doublecortin. By expanding the database as a curated resource, we hope to relate model organism data to clinical findings of pathogenic tubulin variants. Ultimately, we aim to aid researchers in hypothesis generation and design of studies to dissect tubulin function.
Collapse
Affiliation(s)
- Izra Abbaali
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Danny Truong
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Shania Deon Day
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Faliha Mushayeed
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Bhargavi Ganesh
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Nancy Haro-Ramirez
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Juliet Isles
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Hindol Nag
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Catherine Pham
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Priya Shah
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Ishaan Tomar
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Carolina Manel-Romero
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| | - Naomi S. Morrissette
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, United States of America
| |
Collapse
|
27
|
Ren Y, Wang Y, Liu J, Liu T, Yuan L, Wu C, Yang Z, Chen J. X-ray Crystal Structure-Guided Discovery of Novel Indole Analogues as Colchicine-Binding Site Tubulin Inhibitors with Immune-Potentiating and Antitumor Effects against Melanoma. J Med Chem 2023; 66:6697-6714. [PMID: 37145846 DOI: 10.1021/acs.jmedchem.3c00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A series of novel indole analogues were discovered as colchicine-binding site inhibitors of tubulin. Among them, 3a exhibited the highest antiproliferative activity (average IC50 = 4.5 nM), better than colchicine (IC50 = 65.3 nM). The crystal structure of 3a in complex with tubulin was solved by X-ray crystallography, which explained the improved binding affinity of 3a to tubulin and thus its higher anticancer activity (IC50 = 4.5 nM) than the lead compound 12b (IC50 = 32.5 nM). In vivo, 3a (5 mg/kg) displayed significant antitumor efficacy against B16-F10 melanoma with a TGI of 62.96% and enhanced the antitumor efficacy of a small-molecule PD-1/PD-L1 inhibitor NP19 (TGI = 77.85%). Moreover, 3a potentiated the antitumor immunity of NP19 by activating the tumor immune microenvironment, as demonstrated by the increased tumor-infiltrating lymphocytes (TIL). Collectively, this work shows a successful example of crystal structure-guided discovery of a novel tubulin inhibitor 3a as a potential anticancer and immune-potentiating agent.
Collapse
Affiliation(s)
- Yichang Ren
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yuxi Wang
- Targeted Tracer Research and development laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jin Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Ting Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Lin Yuan
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Chengyong Wu
- Targeted Tracer Research and development laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zichao Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
28
|
Carmona B, Marinho HS, Matos CL, Nolasco S, Soares H. Tubulin Post-Translational Modifications: The Elusive Roles of Acetylation. BIOLOGY 2023; 12:biology12040561. [PMID: 37106761 PMCID: PMC10136095 DOI: 10.3390/biology12040561] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
Microtubules (MTs), dynamic polymers of α/β-tubulin heterodimers found in all eukaryotes, are involved in cytoplasm spatial organization, intracellular transport, cell polarity, migration and division, and in cilia biology. MTs functional diversity depends on the differential expression of distinct tubulin isotypes and is amplified by a vast number of different post-translational modifications (PTMs). The addition/removal of PTMs to α- or β-tubulins is catalyzed by specific enzymes and allows combinatory patterns largely enriching the distinct biochemical and biophysical properties of MTs, creating a code read by distinct proteins, including microtubule-associated proteins (MAPs), which allow cellular responses. This review is focused on tubulin-acetylation, whose cellular roles continue to generate debate. We travel through the experimental data pointing to α-tubulin Lys40 acetylation role as being a MT stabilizer and a typical PTM of long lived MTs, to the most recent data, suggesting that Lys40 acetylation enhances MT flexibility and alters the mechanical properties of MTs, preventing MTs from mechanical aging characterized by structural damage. Additionally, we discuss the regulation of tubulin acetyltransferases/desacetylases and their impacts on cell physiology. Finally, we analyze how changes in MT acetylation levels have been found to be a general response to stress and how they are associated with several human pathologies.
Collapse
Affiliation(s)
- Bruno Carmona
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
| | - H Susana Marinho
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Catarina Lopes Matos
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Sofia Nolasco
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Helena Soares
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
| |
Collapse
|
29
|
Novotná Floriančičová K, Baltzis A, Smejkal J, Czerneková M, Kaczmarek Ł, Malý J, Notredame C, Vinopal S. Phylogenetic and functional characterization of water bears (Tardigrada) tubulins. Sci Rep 2023; 13:5194. [PMID: 36997657 PMCID: PMC10063605 DOI: 10.1038/s41598-023-31992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/21/2023] [Indexed: 04/01/2023] Open
Abstract
Tardigrades are microscopic ecdysozoans that can withstand extreme environmental conditions. Several tardigrade species undergo reversible morphological transformations and enter into cryptobiosis, which helps them to survive periods of unfavorable environmental conditions. However, the underlying molecular mechanisms of cryptobiosis are mostly unknown. Tubulins are evolutionarily conserved components of the microtubule cytoskeleton that are crucial in many cellular processes. We hypothesize that microtubules are necessary for the morphological changes associated with successful cryptobiosis. The molecular composition of the microtubule cytoskeleton in tardigrades is unknown. Therefore, we analyzed and characterized tardigrade tubulins and identified 79 tardigrade tubulin sequences in eight taxa. We found three α-, seven β-, one γ-, and one ε-tubulin isoform. To verify in silico identified tardigrade tubulins, we also isolated and sequenced nine out of ten predicted Hypsibius exemplaris tubulins. All tardigrade tubulins were localized as expected when overexpressed in mammalian cultured cells: to the microtubules or to the centrosomes. The presence of a functional ε-tubulin, clearly localized to centrioles, is attractive from a phylogenetic point of view. Although the phylogenetically close Nematoda lost their δ- and ε-tubulins, some groups of Arthropoda still possess them. Thus, our data support the current placement of tardigrades into the Panarthropoda clade.
Collapse
Affiliation(s)
- Kamila Novotná Floriančičová
- Department of Biology, Faculty of Science, J. E. Purkyně University (UJEP), Usti Nad Labem, Czech Republic
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic
| | | | - Jiří Smejkal
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic
| | - Michaela Czerneková
- Department of Biology, Faculty of Science, J. E. Purkyně University (UJEP), Usti Nad Labem, Czech Republic
| | - Łukasz Kaczmarek
- Department of Animal Taxonomy and Ecology, Adam Mickiewicz University in Poznań, Poznań, Poland
| | - Jan Malý
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic
| | - Cedric Notredame
- Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Stanislav Vinopal
- Department of Biology, Faculty of Science, J. E. Purkyně University (UJEP), Usti Nad Labem, Czech Republic.
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic.
| |
Collapse
|
30
|
Kang Y, Pei Y, Qin J, Zhang Y, Duan Y, Yang H, Yao Y, Sun M. Design, synthesis, and biological activity evaluation of novel tubulin polymerization inhibitors based on pyrimidine ring skeletons. Bioorg Med Chem Lett 2023; 84:129195. [PMID: 36828299 DOI: 10.1016/j.bmcl.2023.129195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/02/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023]
Abstract
A library of new pyrimidine analogs was designed and synthesized of these, compound K10 bearing a 1,4‑benzodioxane moiety and 3,4,5‑trimethoxyphenyl group, exhibiting the most potent activity, with IC50 values of 0.07-0.80 μM against four cancer cell lines. Cellular-based mechanism studies elucidated that K10 inhibited microtubule polymerization, blocked the cell cycle at the G2/M phase, and eventually induced apoptosis of HepG2 cells. Additionally, K10 inhibited the migration and invasion of HepG2 cells in a dose-dependent manner. Overall, our work indicates that the tubulin polymerization inhibitor incorporating pyrimidine and the 3,4,5‑trimethoxyphenyl ring may deserve consideration for cancer therapy.
Collapse
Affiliation(s)
- Yingying Kang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuanyuan Pei
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jinling Qin
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China
| | - Hua Yang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yongfang Yao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
31
|
Wranik M, Weinert T, Slavov C, Masini T, Furrer A, Gaillard N, Gioia D, Ferrarotti M, James D, Glover H, Carrillo M, Kekilli D, Stipp R, Skopintsev P, Brünle S, Mühlethaler T, Beale J, Gashi D, Nass K, Ozerov D, Johnson PJM, Cirelli C, Bacellar C, Braun M, Wang M, Dworkowski F, Milne C, Cavalli A, Wachtveitl J, Steinmetz MO, Standfuss J. Watching the release of a photopharmacological drug from tubulin using time-resolved serial crystallography. Nat Commun 2023; 14:903. [PMID: 36807348 PMCID: PMC9936131 DOI: 10.1038/s41467-023-36481-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
The binding and release of ligands from their protein targets is central to fundamental biological processes as well as to drug discovery. Photopharmacology introduces chemical triggers that allow the changing of ligand affinities and thus biological activity by light. Insight into the molecular mechanisms of photopharmacology is largely missing because the relevant transitions during the light-triggered reaction cannot be resolved by conventional structural biology. Using time-resolved serial crystallography at a synchrotron and X-ray free-electron laser, we capture the release of the anti-cancer compound azo-combretastatin A4 and the resulting conformational changes in tubulin. Nine structural snapshots from 1 ns to 100 ms complemented by simulations show how cis-to-trans isomerization of the azobenzene bond leads to a switch in ligand affinity, opening of an exit channel, and collapse of the binding pocket upon ligand release. The resulting global backbone rearrangements are related to the action mechanism of microtubule-destabilizing drugs.
Collapse
Affiliation(s)
- Maximilian Wranik
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Tobias Weinert
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Chavdar Slavov
- Institute of Physical and Theoretical Chemistry, Goethe University, Frankfurt am Main, Germany
| | - Tiziana Masini
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Antonia Furrer
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Natacha Gaillard
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Dario Gioia
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Marco Ferrarotti
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Daniel James
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Hannah Glover
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Melissa Carrillo
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Demet Kekilli
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Robin Stipp
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Petr Skopintsev
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Steffen Brünle
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Tobias Mühlethaler
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - John Beale
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Dardan Gashi
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Karol Nass
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Dmitry Ozerov
- Scientific Computing, Theory and Data, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Philip J M Johnson
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Claudio Cirelli
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Camila Bacellar
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Markus Braun
- Institute of Physical and Theoretical Chemistry, Goethe University, Frankfurt am Main, Germany
| | - Meitian Wang
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Florian Dworkowski
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Chris Milne
- Photon Science Division, Paul Scherrer Institut, 5232, Villigen, Switzerland
| | - Andrea Cavalli
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genova, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Josef Wachtveitl
- Institute of Physical and Theoretical Chemistry, Goethe University, Frankfurt am Main, Germany
| | - Michel O Steinmetz
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland.
- Biozentrum, University of Basel, 4056, Basel, Switzerland.
| | - Jörg Standfuss
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland.
| |
Collapse
|
32
|
Zhang L, Lin X, Yang Z, Jiang L, Hou Q, Xie Z, Li Y, Pei H. The role of microtubules in microalgae: promotion of lipid accumulation and extraction. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2023; 16:7. [PMID: 36635732 PMCID: PMC9837904 DOI: 10.1186/s13068-023-02257-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/01/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND Microtubules in cells are closely related to the growth and metabolism of microalgae. To date, the study of microalgal microtubules has mainly concentrated on revealing the relationship between microtubule depolymerization and synthesis of precursors for flagellar regeneration. While information on the link between microtubule depolymerization and biosynthesis of precursors for complex organic matter (such as lipid, carbohydrate and protein), is still lacking, a better understanding of this could help to achieve a breakthrough in lipid regulation. With the aim of testing the assumption that microtubule disruption could regulate carbon precursors and redirect carbon flow to promote lipid accumulation, Chlorella sorokiniana SDEC-18 was pretreated with different concentrations of oryzalin. RESULTS Strikingly, microalgae that were pretreated with 1.5 mM oryzalin accumulated lipid contents of 41.06%, which was attributed to carbon redistribution induced by microtubule destruction. To promote the growth of microalgae, two-stage cultivation involving microtubule destruction was employed, which resulted in the lipid productivity being 1.44 times higher than that for microalgae with routine single-stage cultivation, as well as yielding a desirable biodiesel quality following from increases in monounsaturated fatty acid (MUFA) content. Furthermore, full extraction of lipid was achieved after only a single extraction step, because microtubule destruction caused removal of cellulose synthase and thereby blocked cellulose biosynthesis. CONCLUSIONS This study provides an important advance towards observation of microtubules in microalgae through immunocolloidal gold techniques combined with TEM. Moreover, the observation of efficient lipid accumulation and increased cell fragility engendered by microtubule destruction has expanded our knowledge of metabolic regulation by microtubules. Finally, two-stage cultivation involving microtubule destruction has established ideal growth, coupling enhanced lipid accumulation and efficient oil extraction; thus gaining advances in both applied and fundamental research in algal biodiesel production.
Collapse
Affiliation(s)
- Lijie Zhang
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China
| | - Xiao Lin
- grid.5335.00000000121885934Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS UK
| | - Zhigang Yang
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China
| | - Liqun Jiang
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China
| | - Qingjie Hou
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China
| | - Zhen Xie
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China
| | - Yizhen Li
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China
| | - Haiyan Pei
- grid.27255.370000 0004 1761 1174School of Environmental Science and Engineering, Shandong University, Qingdao, 266237 China ,grid.8547.e0000 0001 0125 2443Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433 China ,Shandong Provincial Engineering Center on Environmental Science and Technology, Jinan, 250061 China
| |
Collapse
|
33
|
Argirova M, Guncheva M, Momekov G, Cherneva E, Mihaylova R, Rangelov M, Todorova N, Denev P, Anichina K, Mavrova A, Yancheva D. Modulation Effect on Tubulin Polymerization, Cytotoxicity and Antioxidant Activity of 1H-Benzimidazole-2-Yl Hydrazones. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010291. [PMID: 36615483 PMCID: PMC9822270 DOI: 10.3390/molecules28010291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022]
Abstract
1H-benzimidazol-2-yl hydrazones with varying hydroxy and methoxy phenyl moieties were designed. Their effect on tubulin polymerization was evaluated in vitro on porcine tubulin. The compounds elongated the nucleation phase and slowed down the tubulin polymerization comparably to nocodazole. The possible binding modes of the hydrazones with tubulin were explored by molecular docking at the colchicine binding site. The anticancer activity was evaluated against human malignant cell lines MCF-7 and AR-230, as well as against normal fibroblast cells 3T3 and CCL-1. The compounds demonstrated a marked antineoplastic activity in low micromolar concentrations in both screened in vitro tumor models. The most active were the trimethoxy substituted derivative 1i and the positional isomers 1j and 1k, containing hydroxy and methoxy substituents: they showed IC50 similar to the reference podophyllotoxin in both tumor cell lines, accompanied with high selectivity towards the malignantly transformed cells. The compounds exerted moderate to high ability to scavenge peroxyl radicals and certain derivatives-1l containing metha-hydroxy and para-methoxy group, and 1b-e with di/trihydroxy phenyl moiety, revealed HORAC values high or comparable to those of well-known phenolic antioxidants. Thus the 1H-benisimidazol-2-yl hydrazones with hydroxy/methoxy phenyl fragments were recognized as new agents exhibiting promising combined antioxidant and antineoplastic action.
Collapse
Affiliation(s)
- Maria Argirova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Maya Guncheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Georgi Momekov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Emiliya Cherneva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Rositsa Mihaylova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Miroslav Rangelov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Nadezhda Todorova
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Petko Denev
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Kameliya Anichina
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 1756 Sofia, Bulgaria
| | - Anelia Mavrova
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 1756 Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- Correspondence:
| |
Collapse
|
34
|
Natural Taxanes: From Plant Composition to Human Pharmacology and Toxicity. Int J Mol Sci 2022; 23:ijms232415619. [PMID: 36555256 PMCID: PMC9779243 DOI: 10.3390/ijms232415619] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Biologically active taxanes, present in small- to medium-sized evergreen conifers of various Taxus species, are widely used for their antioxidant, antimicrobial and anti-inflammatory effects, but mostly for their antitumour effects used in the treatment of solid tumours of the breast, ovary, lung, bladder, prostate, oesophagus and melanoma. More of the substances found in Taxus plant extracts have medical potential. Therefore, at the beginning of this review, we describe the methods of isolation, identification and determination of taxanes in different plant parts. One of the most important taxanes is paclitaxel, for which we summarize the pharmacokinetic parameters of its different formulations. We also describe toxicological risks during clinical therapy such as hypersensitivity, neurotoxicity, gastrointestinal, cardiovascular, haematological, skin and renal toxicity and toxicity to the respiratory system. Since the effect of the drug-form PTX is enhanced by various Taxus spp. extracts, we summarize published clinical intoxications and all fatal poisonings for the Taxus baccata plant. This showed that, despite their significant use in anticancer treatment, attention should also be focused on the risk of fatal intoxication due to ingestion of extracts from these plants, which are commonly found in our surroundings.
Collapse
|
35
|
Alhammad R. Bioinformatics Identification of TUBB as Potential Prognostic Biomarker for Worse Prognosis in ERα-Positive and Better Prognosis in ERα-Negative Breast Cancer. Diagnostics (Basel) 2022; 12:diagnostics12092067. [PMID: 36140469 PMCID: PMC9498198 DOI: 10.3390/diagnostics12092067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Tubulin β class I gene (TUBB) is highly expressed in various cancers and plays several roles in carcinogenesis. However, the prognostic value of TUBB in breast cancer remains to be investigated. GEPIA and Breast Cancer Gene-Expression Miner were used to explore TUBB expression in breast cancer patients. Kaplan–Meier Plotter was used to assess the relationship between TUBB expression and several prognostic indicators including overall, distant metastasis-free, and relapse-free survival in ERα-positive and ERα-negative breast cancer. The genes that correlate with TUBB in ERα-positive and ERα-negative breast cancer were explored and the pathways were investigated using GSCA. The correlation between TUBB and several gene markers of immune cells was explored using GEPIA. ERα-positive breast cancer patients with increased TUBB showed worse prognosis, possibly through the activation of the TSC/mTOR pathway, whereas ERα-negative breast cancer patients with increased TUBB mRNA showed better prognosis. Significant positive correlations were observed between TUBB and gene markers of immune cells in ERα-positive breast cancer patients, whereas significant negative correlations were observed in ERα-negative breast cancer patients. The analysis revealed that TUBB might be considered as a predictive biomarker for worse prognosis in ERα-positive and better prognosis in ERα-negative breast cancer.
Collapse
Affiliation(s)
- Rashed Alhammad
- Department of Pharmacology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| |
Collapse
|
36
|
Romagnoli R, Oliva P, Prencipe F, Manfredini S, Budassi F, Brancale A, Ferla S, Hamel E, Corallo D, Aveic S, Manfreda L, Mariotto E, Bortolozzi R, Viola G. Design, Synthesis and Biological Investigation of 2-Anilino Triazolopyrimidines as Tubulin Polymerization Inhibitors with Anticancer Activities. Pharmaceuticals (Basel) 2022; 15:1031. [PMID: 36015179 PMCID: PMC9415608 DOI: 10.3390/ph15081031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
A further investigation aiming to generate new potential antitumor agents led us to synthesize a new series of twenty-two compounds characterized by the presence of the 7-(3',4',5'-trimethoxyphenyl)-[1,2,4]triazolo[1,5-a]pyrimidine pharmacophore modified at its 2-position. Among the synthesized compounds, three were significantly more active than the others. These bore the substituents p-toluidino (3d), p-ethylanilino (3h) and 3',4'-dimethylanilino (3f), and these compounds had IC50 values of 30-43, 160-240 and 67-160 nM, respectively, on HeLa, A549 and HT-29 cancer cells. The p-toluidino derivative 3d was the most potent inhibitor of tubulin polymerization (IC50: 0.45 µM) and strongly inhibited the binding of colchicine to tubulin (72% inhibition), with antiproliferative activity superior to CA-4 against A549 and HeLa cancer cell lines. In vitro investigation showed that compound 3d was able to block treated cells in the G2/M phase of the cell cycle and to induce apoptosis following the intrinsic pathway, further confirmed by mitochondrial depolarization and caspase-9 activation. In vivo experiments conducted on the zebrafish model showed good activity of 3d in reducing the mass of a HeLa cell xenograft. These effects occurred at nontoxic concentrations to the animal, indicating that 3d merits further developmental studies.
Collapse
Affiliation(s)
- Romeo Romagnoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Oliva
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Filippo Prencipe
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
| | - Stefano Manfredini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Federica Budassi
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit-An Evotec Company, 37135 Verona, Italy
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Salvatore Ferla
- Faculty of Medicine, Health and Life Science, Swansea University Medical School, Grove Building, Swansea University, Swansea SA2 8PP, UK
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Lorenzo Manfreda
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Elena Mariotto
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Roberta Bortolozzi
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Giampietro Viola
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| |
Collapse
|
37
|
Daume D, Offner T, Hassenklöver T, Manzini I. Patterns of tubb2b Promoter-Driven Fluorescence in the Forebrain of Larval Xenopus laevis. Front Neuroanat 2022; 16:914281. [PMID: 35873659 PMCID: PMC9304554 DOI: 10.3389/fnana.2022.914281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Microtubules are essential components of the cytoskeleton of all eukaryotic cells and consist of α- and β-tubulin heterodimers. Several tissue-specific isotypes of α- and β-tubulins, encoded by distinct genes, have been described in vertebrates. In the African clawed frog (Xenopus laevis), class II β-tubulin (tubb2b) is expressed exclusively in neurons, and its promoter is used to establish different transgenic frog lines. However, a thorough investigation of the expression pattern of tubb2b has not been carried out yet. In this study, we describe the expression of tubb2b-dependent Katushka fluorescence in the forebrain of premetamorphic Xenopus laevis at cellular resolution. To determine the exact location of Katushka-positive neurons in the forebrain nuclei and to verify the extent of neuronal Katushka expression, we used a transgenic frog line and performed several additional antibody stainings. We found tubb2b-dependent fluorescence throughout the Xenopus forebrain, but not in all neurons. In the olfactory bulb, tubb2b-dependent fluorescence is present in axonal projections from the olfactory epithelium, cells in the mitral cell layer, and fibers of the extrabulbar system, but not in interneurons. We also detected tubb2b-dependent fluorescence in parts of the basal ganglia, the amygdaloid complex, the pallium, the optic nerve, the preoptic area, and the hypothalamus. In the diencephalon, tubb2b-dependent fluorescence occurred mainly in the prethalamus and thalamus. As in the olfactory system, not all neurons of these forebrain regions exhibited tubb2b-dependent fluorescence. Together, our results present a detailed overview of the distribution of tubb2b-dependent fluorescence in neurons of the forebrain of larval Xenopus laevis and clearly show that tubb2b-dependent fluorescence cannot be used as a pan-neuronal marker.
Collapse
|
38
|
Discovery of N-benzylarylamide derivatives as novel tubulin polymerization inhibitors capable of activating the Hippo pathway. Eur J Med Chem 2022; 240:114583. [PMID: 35834904 DOI: 10.1016/j.ejmech.2022.114583] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 01/12/2023]
Abstract
Novel N-benzylarylamide saderivatives were designed and synthesized, and their antiproliferative activities were explored. Some of 51 target compounds exhibited potent inhibitory activities against MGC-803, HCT-116 and KYSE450 cells with IC50 values in two-digit nanomolar. Compound I-33 (MY-875) displayed the most potent antiproliferative activities against MGC-803, HCT-116 and KYSE450 cells (IC50 = 0.027, 0.055 and 0.067 μM, respectively) and possessed IC50 values ranging from 0.025 to 0.094 μM against other 11 cancer cell lines. Further mechanism studies indicated that compound I-33 (MY-875) inhibited tubulin polymerization (IC50 = 0.92 μM) by targeting the colchicine bingding site of tubulin. Compound I-33 (MY-875) disrupted the construction of the microtubule networks and affected the mitosis in MGC-803 and SGC-7901 cells. In addition, although it acted as a colchicine binding site inhibitor, compound I-33 (MY-875) also activated the Hippo pathway to promote the phosphorylation status of MST and LATS, resulting in the YAP degradation in MGC-803 and SGC-7901 cells. Due to the degradation of YAP, the expression levels of TAZ and Axl decreased. Because of the dual actions on colchicine binding site and Hippo pathway, compound I-33 (MY-875) dose-dependently inhibited cell colony formatting ability, arrested cells at the G2/M phase and induced cells apoptosis in MGC-803 and SGC-7901 cells. Moreover, compound I-33 (MY-875) could regulate the levels of cell cycle and apoptosis regulatory proteins in MGC-803 and SGC-7901 cells. Furthermore, molecular docking analysis suggested that the hydrogen bond and hydrophobic interactions made compound I-33 (MY-875) well bind into the colchicine binding site of tubulin. Collectively, compound I-33 (MY-875) is a novel anti-gastric cancer agent and deserves to be further investigated for cancer therapy by targeting the colchicine binding site of tubulin and activating the Hippo pathway.
Collapse
|
39
|
Alpízar-Pedraza D, Veulens ADLN, Araujo EC, Piloto-Ferrer J, Sánchez-Lamar Á. Microtubules destabilizing agents binding sites in tubulin. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
de la Roche NM, Mühlethaler T, Di Martino RMC, Ortega JA, Gioia D, Roy B, Prota AE, Steinmetz MO, Cavalli A. Novel fragment-derived colchicine-site binders as microtubule-destabilizing agents. Eur J Med Chem 2022; 241:114614. [DOI: 10.1016/j.ejmech.2022.114614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022]
|
41
|
Shemesh A, Ginsburg A, Dharan R, Levi-Kalisman Y, Ringel I, Raviv U. Mechanism of Tubulin Oligomers and Single-Ring Disassembly Catastrophe. J Phys Chem Lett 2022; 13:5246-5252. [PMID: 35671351 PMCID: PMC9208022 DOI: 10.1021/acs.jpclett.2c00947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Cold tubulin dimers coexist with tubulin oligomers and single rings. These structures are involved in microtubule assembly; however, their dynamics are poorly understood. Using state-of-the-art solution synchrotron time-resolved small-angle X-ray scattering, we discovered a disassembly catastrophe (half-life of ∼0.1 s) of tubulin rings and oligomers upon dilution or addition of guanosine triphosphate. A slower disassembly (half-life of ∼38 s) was observed following an increase in temperature. Our analysis showed that the assembly and disassembly processes were consistent with an isodesmic mechanism, involving a sequence of reversible reactions in which dimers were rapidly added or removed one at a time, terminated by a 2 order-of-magnitude slower ring-closing/opening step. We revealed how assembly conditions varied the mass fraction of tubulin in each of the coexisting structures, the rate constants, and the standard Helmholtz free energies for closing a ring and for longitudinal dimer-dimer associations.
Collapse
Affiliation(s)
- Asaf Shemesh
- Institute
of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- Center
for Nanoscience and Nanotechnology, The
Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Avi Ginsburg
- Institute
of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Raviv Dharan
- Institute
of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yael Levi-Kalisman
- Center
for Nanoscience and Nanotechnology, The
Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- Institute
of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Israel Ringel
- Institute
for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Uri Raviv
- Institute
of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- Center
for Nanoscience and Nanotechnology, The
Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
42
|
Oliva P, Romagnoli R, Cacciari B, Manfredini S, Padroni C, Brancale A, Ferla S, Hamel E, Corallo D, Aveic S, Milan N, Mariotto E, Viola G, Bortolozzi R. Synthesis and Biological Evaluation of Highly Active 7-Anilino Triazolopyrimidines as Potent Antimicrotubule Agents. Pharmaceutics 2022; 14:1191. [PMID: 35745764 PMCID: PMC9230136 DOI: 10.3390/pharmaceutics14061191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
Two different series of fifty-two compounds, based on 3',4',5'-trimethoxyaniline (7a-ad) and variably substituted anilines (8a-v) at the 7-position of the 2-substituted-[1,2,4]triazolo [1,5-a]pyrimidine nucleus, had moderate to potent antiproliferative activity against A549, MDA-MB-231, HeLa, HT-29 and Jurkat cancer cell lines. All derivatives with a common 3-phenylpropylamino moiety at the 2-position of the triazolopyrimidine scaffold and different halogen-substituted anilines at its 7-position, corresponding to 4'-fluoroaniline (8q), 4'-fluoro-3'-chloroaniline (8r), 4'-chloroaniline (8s) and 4'-bromoaniline (8u), displayed the greatest antiproliferative activity with mean IC50's of 83, 101, 91 and 83 nM, respectively. These four compounds inhibited tubulin polymerization about 2-fold more potently than combretastatin A-4 (CA-4), and their activities as inhibitors of [3H]colchicine binding to tubulin were similar to that of CA-4. These data underlined that the 3',4',5'-trimethoxyanilino moiety at the 7-position of the [1,2,4]triazolo [1,5-a]pyrimidine system, which characterized compounds 7a-ad, was not essential for maintaining potent antiproliferative and antitubulin activities. Compounds 8q and 8r had high selectivity against cancer cells, and their interaction with tubulin led to the accumulation of HeLa cells in the G2/M phase of the cell cycle and to apoptotic cell death through the mitochondrial pathway. Finally, compound 8q significantly inhibited HeLa cell growth in zebrafish embryos.
Collapse
Affiliation(s)
- Paola Oliva
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (P.O.); (B.C.)
| | - Romeo Romagnoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (P.O.); (B.C.)
| | - Barbara Cacciari
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (P.O.); (B.C.)
| | - Stefano Manfredini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Padroni
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit—An Evotec Company, Via A. Fleming, 37135 Verona, Italy;
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK;
| | - Salvatore Ferla
- Swansea University Medical School, Institute of Life Sciences 2, Swansea University, Swansea SA2 8PP, UK;
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA;
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy; (D.C.); (S.A.)
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy; (D.C.); (S.A.)
| | - Noemi Milan
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
| | - Elena Mariotto
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy
| | - Giampietro Viola
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy
| | - Roberta Bortolozzi
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy
| |
Collapse
|
43
|
Seo D, Gammon DB. Manipulation of Host Microtubule Networks by Viral Microtubule-Associated Proteins. Viruses 2022; 14:v14050979. [PMID: 35632720 PMCID: PMC9147350 DOI: 10.3390/v14050979] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/19/2022] Open
Abstract
Diverse DNA and RNA viruses utilize cytoskeletal networks to efficiently enter, replicate, and exit the host cell, while evading host immune responses. It is well established that the microtubule (MT) network is commonly hijacked by viruses to traffic to sites of replication after entry and to promote egress from the cell. However, mounting evidence suggests that the MT network is also a key regulator of host immune responses to infection. At the same time, viruses have acquired mechanisms to manipulate and/or usurp MT networks to evade these immune responses. Central to most interactions of viruses with the MT network are virally encoded microtubule-associated proteins (MAPs) that bind to MTs directly or indirectly. These MAPs associate with MTs and other viral or cellular MAPs to regulate various aspects of the MT network, including MT dynamics, MT-dependent transport via motor proteins such as kinesins and dyneins, and MT-dependent regulation of innate immune responses. In this review, we examine how viral MAP interactions with the MT network facilitate viral replication and immune evasion.
Collapse
|
44
|
Jones JH, Minshall RD. Endothelial Transcytosis in Acute Lung Injury: Emerging Mechanisms and Therapeutic Approaches. Front Physiol 2022; 13:828093. [PMID: 35431977 PMCID: PMC9008570 DOI: 10.3389/fphys.2022.828093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury (ALI) is characterized by widespread inflammation which in its severe form, Acute Respiratory Distress Syndrome (ARDS), leads to compromise in respiration causing hypoxemia and death in a substantial number of affected individuals. Loss of endothelial barrier integrity, pneumocyte necrosis, and circulating leukocyte recruitment into the injured lung are recognized mechanisms that contribute to the progression of ALI/ARDS. Additionally, damage to the pulmonary microvasculature by Gram-negative and positive bacteria or viruses (e.g., Escherichia coli, SARS-Cov-2) leads to increased protein and fluid permeability and interstitial edema, further impairing lung function. While most of the vascular leakage is attributed to loss of inter-endothelial junctional integrity, studies in animal models suggest that transendothelial transport of protein through caveolar vesicles, known as transcytosis, occurs in the early phase of ALI/ARDS. Here, we discuss the role of transcytosis in healthy and injured endothelium and highlight recent studies that have contributed to our understanding of the process during ALI/ARDS. We also cover potential approaches that utilize caveolar transport to deliver therapeutics to the lungs which may prevent further injury or improve recovery.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,*Correspondence: Richard D. Minshall,
| |
Collapse
|
45
|
Krajka V, Vulinovic F, Genova M, Tanzer K, Jijumon AS, Bodakuntla S, Tennstedt S, Mueller-Fielitz H, Meier B, Janke C, Klein C, Rakovic A. H-ABC- and dystonia-causing TUBB4A mutations show distinct pathogenic effects. SCIENCE ADVANCES 2022; 8:eabj9229. [PMID: 35275727 PMCID: PMC8916731 DOI: 10.1126/sciadv.abj9229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 01/22/2022] [Indexed: 06/14/2023]
Abstract
Mutations in the brain-specific β-tubulin 4A (TUBB4A) gene cause a broad spectrum of diseases, ranging from dystonia (DYT-TUBB4A) to hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC). Currently, the mechanisms of how TUBB4A variants lead to this pleiotropic manifestation remain elusive. Here, we investigated whether TUBB4A mutations causing either DYT-TUBB4A (p.R2G and p.Q424H) or H-ABC (p.R2W and p.D249N) exhibit differential effects at the molecular and cellular levels. Using live-cell imaging of disease-relevant oligodendrocytes and total internal reflection fluorescence microscopy of whole-cell lysates, we observed divergent impact on microtubule polymerization and microtubule integration, partially reflecting the observed pleiotropy. Moreover, in silico simulations demonstrated that the mutants rarely adopted a straight heterodimer conformation in contrast to wild type. In conclusion, for most of the examined variants, we deciphered potential molecular disease mechanisms that may lead to the diverse clinical manifestations and phenotype severity across and within each TUBB4A-related disease.
Collapse
Affiliation(s)
- Victor Krajka
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
- Institute of Microtechnology (IMT), Technische Universität Braunschweig, Braunschweig 38124, Germany
| | - Franca Vulinovic
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Mariya Genova
- Institut Curie, Université PSL, CNRS UMR3348, 91401 Orsay, France
- Université Paris-Saclay, CNRS UMR3348, 91401 Orsay, France
| | - Kerstin Tanzer
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - A. S. Jijumon
- Institut Curie, Université PSL, CNRS UMR3348, 91401 Orsay, France
- Université Paris-Saclay, CNRS UMR3348, 91401 Orsay, France
| | - Satish Bodakuntla
- Institut Curie, Université PSL, CNRS UMR3348, 91401 Orsay, France
- Université Paris-Saclay, CNRS UMR3348, 91401 Orsay, France
| | - Stephanie Tennstedt
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
- University Heart Center Lübeck, 23562 Lübeck, Germany
| | - Helge Mueller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Britta Meier
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Carsten Janke
- Institut Curie, Université PSL, CNRS UMR3348, 91401 Orsay, France
- Université Paris-Saclay, CNRS UMR3348, 91401 Orsay, France
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Aleksandar Rakovic
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| |
Collapse
|
46
|
Li Y, Liu Y, Zhu Z, Yan W, Zhang C, Yang Z, Bai P, Tang M, Shi M, He W, Fu S, Liu J, Han K, Li J, Xie L, Ye H, Yang J, Chen L. Structure-Based Design and Synthesis of N-Substituted 3-Amino-β-Carboline Derivatives as Potent αβ-Tubulin Degradation Agents. J Med Chem 2022; 65:2675-2693. [PMID: 35084853 DOI: 10.1021/acs.jmedchem.1c02159] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
So far, relatively few small molecules have been reported to promote tubulin degradation. Our previous studies have found that compound 2, a noncovalent colchicine-site ligand, was capable of promoting αβ-tubulin degradation. To further improve its antiproliferative activity, 66 derivatives or analogues of 2 were designed and synthesized based on 2-tubulin cocrystal structure. Among them, 12b displayed nanomolar potency against a variety of tumor cells, including paclitaxel- and adriamycin-resistant cell lines. 12b binds to the colchicine site and promotes αβ-tubulin degradation in a concentration-dependent manner via the ubiquitin-proteasome pathway. The X-ray crystal structure revealed that 12b binds in a similar manner as 2, but there is a slight conformation change of the B ring, which resulted in better interaction of 12b with surrounding residues. 12b effectively suppressed tumor growth at an i.v. dose of 40 mg/kg (3 times a week) on both A2780S (paclitaxel-sensitive) and A2780T (paclitaxel-resistant) ovarian xenograft models, with respective TGIs of 92.42 and 79.75% without obvious side effects, supporting its potential utility as a tumor-therapeutic compound.
Collapse
Affiliation(s)
- Yong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Chufeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Peng Bai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Wen He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Suhong Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jiang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Kai Han
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jiewen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Lixin Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| |
Collapse
|
47
|
Saunders HAJ, Johnson-Schlitz DM, Jenkins BV, Volkert PJ, Yang SZ, Wildonger J. Acetylated α-tubulin K394 regulates microtubule stability to shape the growth of axon terminals. Curr Biol 2022; 32:614-630.e5. [PMID: 35081332 PMCID: PMC8843987 DOI: 10.1016/j.cub.2021.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/19/2021] [Accepted: 12/07/2021] [Indexed: 02/09/2023]
Abstract
Microtubules are essential to neuron shape and function. Acetylation of tubulin has the potential to directly tune the behavior and function of microtubules in cells. Although proteomic studies have identified several acetylation sites in α-tubulin, the effects of acetylation at these sites remains largely unknown. This includes the highly conserved residue lysine 394 (K394), which is located at the αβ-tubulin dimer interface. Using a fly model, we show that α-tubulin K394 is acetylated in the nervous system and is an essential residue. We found that an acetylation-blocking mutation in endogenous α-tubulin, K394R, perturbs the synaptic morphogenesis of motoneurons and reduces microtubule stability. Intriguingly, the K394R mutation has opposite effects on the growth of two functionally and morphologically distinct motoneurons, revealing neuron-type-specific responses when microtubule stability is altered. Eliminating the deacetylase HDAC6 increases K394 acetylation, and the over-expression of HDAC6 reduces microtubule stability similar to the K394R mutant. Thus, our findings implicate α-tubulin K394 and its acetylation in the regulation of microtubule stability and suggest that HDAC6 regulates K394 acetylation during synaptic morphogenesis.
Collapse
Affiliation(s)
- Harriet A. J. Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Dena M. Johnson-Schlitz
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Brian V. Jenkins
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Peter J. Volkert
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Biochemistry Scholars Program, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Sihui Z. Yang
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Cellular & Molecular Biology Graduate Program, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Current address: Pediatrics Department and Biological Sciences Division, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA,Lead and author for correspondence:
| |
Collapse
|
48
|
Bolanos-Garcia VM. On the Regulation of Mitosis by the Kinetochore, a Macromolecular Complex and Organising Hub of Eukaryotic Organisms. Subcell Biochem 2022; 99:235-267. [PMID: 36151378 DOI: 10.1007/978-3-031-00793-4_7] [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: 06/16/2023]
Abstract
The kinetochore is the multiprotein complex of eukaryotic organisms that is assembled on mitotic or meiotic centromeres to connect centromeric DNA with microtubules. Its function involves the coordinated action of more than 100 different proteins. The kinetochore acts as an organiser hub that establishes physical connections with microtubules and centromere-associated proteins and recruits central protein components of the spindle assembly checkpoint (SAC), an evolutionarily conserved surveillance mechanism of eukaryotic organisms that detects unattached kinetochores and destabilises incorrect kinetochore-microtubule attachments. The molecular communication between the kinetochore and the SAC is highly dynamic and tightly regulated to ensure that cells can progress towards anaphase until each chromosome is properly bi-oriented on the mitotic spindle. This is achieved through an interplay of highly cooperative interactions and concerted phosphorylation/dephosphorylation events that are organised in time and space.This contribution discusses our current understanding of the function, structure and regulation of the kinetochore, in particular, how its communication with the SAC results in the amplification of specific signals to exquisitely control the eukaryotic cell cycle. This contribution also addresses recent advances in machine learning approaches, cell imaging and proteomics techniques that have enhanced our understanding of the molecular mechanisms that ensure the high fidelity and timely segregation of the genetic material every time a cell divides as well as the current challenges in the study of this fascinating molecular machine.
Collapse
Affiliation(s)
- Victor M Bolanos-Garcia
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK.
| |
Collapse
|
49
|
Gaillard N, Sharma A, Abbaali I, Liu T, Shilliday F, Cook AD, Ehrhard V, Bangera M, Roberts AJ, Moores CA, Morrissette N, Steinmetz MO. Inhibiting parasite proliferation using a rationally designed anti-tubulin agent. EMBO Mol Med 2021; 13:e13818. [PMID: 34661376 PMCID: PMC8573600 DOI: 10.15252/emmm.202013818] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 09/17/2021] [Accepted: 09/29/2021] [Indexed: 11/09/2022] Open
Abstract
Infectious diseases caused by apicomplexan parasites remain a global public health threat. The presence of multiple ligand-binding sites in tubulin makes this protein an attractive target for anti-parasite drug discovery. However, despite remarkable successes as anti-cancer agents, the rational development of protozoan parasite-specific tubulin drugs has been hindered by a lack of structural and biochemical information on protozoan tubulins. Here, we present atomic structures for a protozoan tubulin and microtubule and delineate the architectures of apicomplexan tubulin drug-binding sites. Based on this information, we rationally designed the parasite-specific tubulin inhibitor parabulin and show that it inhibits growth of parasites while displaying no effects on human cells. Our work presents for the first time the rational design of a species-specific tubulin drug providing a framework to exploit structural differences between human and protozoa tubulin variants enabling the development of much-needed, novel parasite inhibitors.
Collapse
Affiliation(s)
- Natacha Gaillard
- Laboratory of Biomolecular ResearchDivision of Biology and ChemistryPaul Scherrer InstitutVilligenSwitzerland
| | - Ashwani Sharma
- Laboratory of Biomolecular ResearchDivision of Biology and ChemistryPaul Scherrer InstitutVilligenSwitzerland
| | - Izra Abbaali
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Tianyang Liu
- Institute of Structural and Molecular BiologyBirkbeck, University of LondonLondonUK
| | - Fiona Shilliday
- Institute of Structural and Molecular BiologyBirkbeck, University of LondonLondonUK
| | - Alexander D Cook
- Institute of Structural and Molecular BiologyBirkbeck, University of LondonLondonUK
| | - Valentin Ehrhard
- Laboratory of Biomolecular ResearchDivision of Biology and ChemistryPaul Scherrer InstitutVilligenSwitzerland
| | - Mamata Bangera
- Institute of Structural and Molecular BiologyBirkbeck, University of LondonLondonUK
| | - Anthony J Roberts
- Institute of Structural and Molecular BiologyBirkbeck, University of LondonLondonUK
| | - Carolyn A Moores
- Institute of Structural and Molecular BiologyBirkbeck, University of LondonLondonUK
| | - Naomi Morrissette
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Michel O Steinmetz
- Laboratory of Biomolecular ResearchDivision of Biology and ChemistryPaul Scherrer InstitutVilligenSwitzerland
- Biozentrum University of BaselBaselSwitzerland
| |
Collapse
|
50
|
Repurposing of the ALK Inhibitor Crizotinib for Acute Leukemia and Multiple Myeloma Cells. Pharmaceuticals (Basel) 2021; 14:ph14111126. [PMID: 34832908 PMCID: PMC8617756 DOI: 10.3390/ph14111126] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Crizotinib was a first generation of ALK tyrosine kinase inhibitor approved for the treatment of ALK-positive non-small-cell lung carcinoma (NSCLC) patients. COMPARE and cluster analyses of transcriptomic data of the NCI cell line panel indicated that genes with different cellular functions regulated the sensitivity or resistance of cancer cells to crizotinib. Transcription factor binding motif analyses in gene promoters divulged two transcription factors possibly regulating the expression of these genes, i.e., RXRA and GATA1, which are important for leukemia and erythroid development, respectively. COMPARE analyses also implied that cell lines of various cancer types displayed varying degrees of sensitivity to crizotinib. Unexpectedly, leukemia but not lung cancer cells were the most sensitive cells among the different types of NCI cancer cell lines. Re-examining this result in another panel of cell lines indeed revealed that crizotinib exhibited potent cytotoxicity towards acute myeloid leukemia and multiple myeloma cells. P-glycoprotein-overexpressing CEM/ADR5000 leukemia cells were cross-resistant to crizotinib. NCI-H929 multiple myeloma cells were the most sensitive cells. Hence, we evaluated the mode of action of crizotinib on these cells. Although crizotinib is a TKI, it showed highest correlation rates with DNA topoisomerase II inhibitors and tubulin inhibitors. The altered gene expression profiles after crizotinib treatment predicted several networks, where TOP2A and genes related to cell cycle were downregulated. Cell cycle analyses showed that cells incubated with crizotinib for 24 h accumulated in the G2M phase. Crizotinib also increased the number of p-H3(Ser10)-positive NCI-H929 cells illustrating crizotinib's ability to prevent mitotic exit. However, cells accumulated in the sub-G0G1 fraction with longer incubation periods, indicating apoptosis induction. Additionally, crizotinib disassembled the tubulin network of U2OS cells expressing an α-tubulin-GFP fusion protein, preventing migration of cancer cells. This result was verified by in vitro tubulin polymerization assays. In silico molecular docking also revealed a strong binding affinity of crizotinib to the colchicine and Vinca alkaloid binding sites. Taken together, these results demonstrate that crizotinib destabilized microtubules. Additionally, the decatenation assay showed that crizotinib partwise inhibited the catalytic activity of DNA topoisomerase II. In conclusion, crizotinib exerted kinase-independent cytotoxic effects through the dual inhibition of tubulin polymerization and topoisomerase II and might be used to treat not only NSCLC but also multiple myeloma.
Collapse
|