1
|
Chaudhuri R, Dayal N, Kaiser J, Mohallem R, Brauer NR, Yeboah KS, Aryal UK, Sintim HO. Morpholino nicotinamide analogs of ponatinib, dual MNK, p70S6K inhibitors, display efficacy against lung and breast cancers. Bioorg Chem 2025; 159:108298. [PMID: 40081260 DOI: 10.1016/j.bioorg.2025.108298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025]
Abstract
Therapeutic options for aggressive cancer types such as breast and lung remain limited; disease relapse and death occur in 30-60% of non-small cell lung cancer (NSCLC) patients, whereas in triple-negative breast cancer or TNBC, recurrence-free survival occurs in less than 30% patients. The kinases, MNK and p70S6K have been proposed as targets for the potential treatment of breast cancer (BC) and lung cancer but currently, no drug that was purposely designed to inhibit these kinases have been approved by the FDA for the treatment of BC or NSCLC. In this study, we have identified HSND80 (a morpholino nicotinamide analog of ponatinib) as a potent MNK/p70S6K inhibitor that has excellent activity against TNBC and NSCLC cell lines. HSND80 has a longer target residence time (τ) of 45 mins and 58 mins against MNK1 and MNK2 respectively, compared to τ of eFT508 (tomivosertib) against MNK1 and MNK2 (τ = 1 min and 5 min, respectively). Molecular dynamics simulation was used to provide some insights into the binding of HSND80 to MNK and p70S6K kinases. Western blotting analysis and phosphoproteomics analysis of the TNBC cell line, MDA-MB-231, revealed that phosphorylations of elF4E (MNK target) and elF4B and S6 (p70S6K targets) were reduced upon compound treatment, which is in line with the proposed mechanism of action; dual MNK/p70S6K targeting. HSND80 could be dosed orally at 15 and 30 mg/kg and at such doses, could reduce tumor volume in a syngeneic NSCLC mouse model.
Collapse
Affiliation(s)
- Riddhi Chaudhuri
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Neetu Dayal
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Joshua Kaiser
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Rodrigo Mohallem
- Department of Comparative Pathobiology, Purdue University, 1203 W State Street, West Lafayette, IN 47907, USA
| | - Nickolas R Brauer
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Kofi Simpa Yeboah
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Uma K Aryal
- Department of Comparative Pathobiology, Purdue University, 1203 W State Street, West Lafayette, IN 47907, USA; Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, 1203 W State Street, West Lafayette, IN 47907, USA
| | - Herman O Sintim
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA; Purdue Institute for Cancer Research, Purdue University, 201 S. University Street, West Lafayette, IN 47907, USA; Department of Chemistry and Biochemistry, University of Notre Dame, 305A McCourtney Hall, Notre Dame, IN 46556, USA; Mike and Josie Harper Cancer Research Institute, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA.
| |
Collapse
|
2
|
Fatma M, Parveen S, Mir SS. Unraveling the kinase code: Role of protein kinase in lung cancer pathogenesis and therapeutic strategies. Biochim Biophys Acta Rev Cancer 2025; 1880:189309. [PMID: 40169080 DOI: 10.1016/j.bbcan.2025.189309] [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: 08/09/2024] [Revised: 02/05/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
Lung cancer is a prominent cause of cancer-related deaths globally, prompting exploration into the molecular pathways governing cancer cell signaling. Recent insights highlight the critical role of kinases in carcinogenesis and metastasis, particularly in non-small cell lung cancer (NSCLC), where protein kinases significantly contribute to drug resistance. These diverse enzymes catalyze protein phosphorylation and are implicated in cancer through misregulated expression, amplification, aberrant phosphorylation, mutations, and chromosomal translocations. Amplifications of kinases serve as important diagnostic, prognostic, and predictive biomarkers across various cancers. Notably, the Phosphatidylinositol 3-kinase (PI3K)/AKT pathway is crucial for the survival and proliferation of tumor cells. Novel therapeutic approaches are being explored to precisely target these pathways. Peptide-based therapies offer specificity and reduced toxicity compared to conventional treatments, while gene therapy targets abnormal genetic expressions. Advances in nanotechnology and CRISPR/Cas9 systems enhance gene delivery methods, holding promise for targeting specific molecular pathways in lung cancer treatment and minimizing systemic toxicity.
Collapse
Affiliation(s)
- Mariyam Fatma
- Molecular Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary Research-4 (ICEIR-4) Integral University, Kursi Road, Lucknow 226026, India; Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow 226026, India
| | - Sana Parveen
- Molecular Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary Research-4 (ICEIR-4) Integral University, Kursi Road, Lucknow 226026, India; Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow 226026, India
| | - Snober S Mir
- Molecular Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary Research-4 (ICEIR-4) Integral University, Kursi Road, Lucknow 226026, India; Department of Biosciences, Faculty of Science, Integral University, Kursi Road, Lucknow 226026, India.
| |
Collapse
|
3
|
Navarro AM, Alonso M, Martínez-Pérez E, Lazar T, Gibson TJ, Iserte JA, Tompa P, Marino-Buslje C. Unveiling the Complexity of cis-Regulation Mechanisms in Kinases: A Comprehensive Analysis. Proteins 2025; 93:575-587. [PMID: 39366918 DOI: 10.1002/prot.26751] [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: 04/26/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 10/06/2024]
Abstract
Protein cis-regulatory elements (CREs) are regions that modulate the activity of a protein through intramolecular interactions. Kinases, pivotal enzymes in numerous biological processes, often undergo regulatory control via inhibitory interactions in cis. This study delves into the mechanisms of cis regulation in kinases mediated by CREs, employing a combined structural and sequence analysis. To accomplish this, we curated an extensive dataset of kinases featuring annotated CREs, organized into homolog families through multiple sequence alignments. Key molecular attributes, including disorder and secondary structure content, active and ATP-binding sites, post-translational modifications, and disease-associated mutations, were systematically mapped onto all sequences. Additionally, we explored the potential for conformational changes between active and inactive states. Finally, we explored the presence of these kinases within membraneless organelles and elucidated their functional roles therein. CREs display a continuum of structures, ranging from short disordered stretches to fully folded domains. The adaptability demonstrated by CREs in achieving the common goal of kinase inhibition spans from direct autoinhibitory interaction with the active site within the kinase domain, to CREs binding to an alternative site, inducing allosteric regulation revealing distinct types of inhibitory mechanisms, which we exemplify by archetypical representative systems. While this study provides a systematic approach to comprehend kinase CREs, further experimental investigations are imperative to unravel the complexity within distinct kinase families. The insights gleaned from this research lay the foundation for future studies aiming to decipher the molecular basis of kinase dysregulation, and explore potential therapeutic interventions.
Collapse
Affiliation(s)
- Alvaro M Navarro
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Macarena Alonso
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | | | - Tamas Lazar
- VIB-VUB Center for Structural Biology, Flanders Institute for Biotechnology (VIB), Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Javier A Iserte
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Peter Tompa
- VIB-VUB Center for Structural Biology, Flanders Institute for Biotechnology (VIB), Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
- Research Centre for Natural Sciences, Hungarian Research Network, Institute of Enzymology, Budapest, Hungary
| | | |
Collapse
|
4
|
Che X, Zhao Y, Xu Z, Hu Y, Ren A, Wu C, Yang J. Unlocking the Potential of l-α-Glycerylphosphorylcholine: From Metabolic Pathways to Therapeutic Applications. Nutr Rev 2025:nuaf008. [PMID: 40036805 DOI: 10.1093/nutrit/nuaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
l-α-Glycerylphosphorylcholine (GPC), also known as choline alphoscerate or α-glycerophosphorylcholine, serves as both a pharmaceutical product and a dietary supplement. Through its metabolic pathways, GPC acts as the precursor not only of choline and acetylcholine but also of various phospholipids. Extensive preclinical and clinical evidence demonstrates that GPC effectively alleviates cognitive impairment associated with Alzheimer's disease, vascular dementia, cerebral ischemia, stress, and epilepsy, among other conditions. Additionally, GPC has beneficial effects on such conditions and measures as ischemic/hypoxic conditions, ionizing radiation-induced damage, exercise performance, growth hormone release, and liver damage. As well as facilitating cholinergic neurotransmission, evidence also indicates GPC, among other activities, also can promote γ-aminobutyric acid release, enhance protein kinase C activity, facilitate hippocampal neurogenesis, upregulate neurotrophic factors, and inhibit inflammation. In preclinical studies, results indicate that GPC is not genotoxic in vitro or in vivo. Extensive human studies indicate GPC causes no severe adverse effects. Possible risks of atherosclerosis and stroke await necessary validation. In this review, the GPC-related metabolic pathways, pharmacological effects, mechanisms of action, and safety evaluation are discussed with the aim of providing a comprehensive understanding of GPC.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yang Zhao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhongtian Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yue Hu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Aoxin Ren
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
5
|
Di Stefano M, Piazza L, Poles C, Galati S, Granchi C, Giordano A, Campisi L, Macchia M, Poli G, Tuccinardi T. KinasePred: A Computational Tool for Small-Molecule Kinase Target Prediction. Int J Mol Sci 2025; 26:2157. [PMID: 40076779 PMCID: PMC11900317 DOI: 10.3390/ijms26052157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Protein kinases are key regulators of cellular processes and critical therapeutic targets in diseases like cancer, making them a focal point for drug discovery efforts. In this context, we developed KinasePred, a robust computational workflow that combines machine learning and explainable artificial intelligence to predict the kinase activity of small molecules while providing detailed insights into the structural features driving ligand-target interactions. Our kinase-family predictive tool demonstrated significant performance, validated through virtual screening, where it successfully identified six kinase inhibitors. Target-focused operational models were subsequently developed to refine target-specific predictions, enabling the identification of molecular determinants of kinase selectivity. This integrated framework not only accelerates the screening and identification of kinase-targeting compounds but also supports broader applications in target identification, polypharmacology studies, and off-target effect analysis, providing a versatile tool for streamlining the drug discovery process.
Collapse
Affiliation(s)
- Miriana Di Stefano
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Lisa Piazza
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Clarissa Poles
- Telethon Institute of Genetics and Medicine, 80078 Naples, Italy;
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale (SSM, School of Advanced Studies), 80078 Naples, Italy
| | - Salvatore Galati
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Luca Campisi
- Flashtox srl, Via Tosco Romagnola 136, 56025 Pontedera, Italy;
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| |
Collapse
|
6
|
Gentry ZO, McAteer OD, Hamad JL, Moran JA, Kim JT, Marsden MD, Zack JA, Wender PA. Synthesis and preclinical evaluation of tigilanol tiglate analogs as latency-reversing agents for the eradication of HIV. SCIENCE ADVANCES 2025; 11:eads1911. [PMID: 39854456 PMCID: PMC11778240 DOI: 10.1126/sciadv.ads1911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025]
Abstract
Tigilanol tiglate (EBC-46) is a selective modulator of protein kinase C (PKC) isoforms that is Food and Drug Administration (FDA) approved for the treatment of mast cell tumors in canines with up to an 88% cure rate. Recently, it has been FDA approved for the treatment of soft tissue sarcomas in humans. The role of EBC-46 and, especially, its analogs in efforts to eradicate HIV, treat neurological and cardiovascular disorders, or enhance antigen density in antigen-targeted chimeric antigen receptor-T cell and chimeric antigen receptor-natural killer cell immunotherapies has not been reported. Enabled by our previously reported scalable synthesis of EBC-46, we report herein the systematic design, synthesis, and evaluation of EBC-46 analogs, including those inaccessible from the natural source and their PKC affinities, ability to translocate PKC, nuclear factor κB activity, and efficacy in reversing HIV latency in Jurkat-Latency cells. Leading analogs show exceptional PKC affinities, isoform selectivities, and functional activities, serving as promising candidates for therapeutic applications.
Collapse
Affiliation(s)
- Zachary O. Gentry
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Owen D. McAteer
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jennifer L. Hamad
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jose A. Moran
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Jocelyn T. Kim
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew D. Marsden
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
- Department of Medicine (Division of Infectious Diseases), School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Jerome A. Zack
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
7
|
Ion GND, Nitulescu GM, Mihai DP. Machine Learning-Assisted Drug Repurposing Framework for Discovery of Aurora Kinase B Inhibitors. Pharmaceuticals (Basel) 2024; 18:13. [PMID: 39861075 PMCID: PMC11768374 DOI: 10.3390/ph18010013] [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: 12/09/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Aurora kinase B (AurB) is a pivotal regulator of mitosis, making it a compelling target for cancer therapy. Despite significant advances in protein kinase inhibitor development, there are currently no AurB inhibitors readily available for therapeutic use. Methods: This study introduces a machine learning-assisted drug repurposing framework integrating quantitative structure-activity relationship (QSAR) modeling, molecular fingerprints-based classification, molecular docking, and molecular dynamics (MD) simulations. Using this pipeline, we analyzed 4680 investigational and approved drugs from DrugBank database. Results: The machine learning models trained for drug repurposing showed satisfying performance and yielded the identification of saredutant, montelukast, and canertinib as potential AurB inhibitors. The candidates demonstrated strong binding energies, key molecular interactions with critical residues (e.g., Phe88, Glu161), and stable MD trajectories, particularly saredutant, a neurokinin-2 (NK2) antagonist. Conclusions: Beyond identifying potential AurB inhibitors, this study highlights an integrated methodology that can be applied to other challenging drug targets.
Collapse
Affiliation(s)
| | - George Mihai Nitulescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (G.N.D.I.); (D.P.M.)
| | | |
Collapse
|
8
|
Gómez-García A, Acuña Jiménez DA, Zamora WJ, Barazorda-Ccahuana HL, Chávez-Fumagalli MÁ, Valli M, Andricopulo AD, Bolzani VDS, Olmedo DA, Solís PN, Núñez MJ, Rodríguez Pérez JR, Valencia Sánchez HA, Cortés Hernández HF, Mosquera Martinez OM, Medina-Franco JL. Latin American Natural Product Database (LANaPDB): An Update. J Chem Inf Model 2024; 64:8495-8509. [PMID: 39503579 PMCID: PMC11600509 DOI: 10.1021/acs.jcim.4c01560] [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: 08/29/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024]
Abstract
Natural product (NP) databases are crucial tools in computer-aided drug design (CADD). Over the past decade, there has been a worldwide effort to assemble information regarding natural products (NPs) isolated and characterized in certain geographical regions. In 2023, it was published LANaPDB, and to our knowledge, this is the first attempt to gather and standardize all the NP databases of Latin America. Herein, we present and analyze in detail the contents of an updated version of LANaPDB, which includes 619 newly added compounds from Colombia, Costa Rica, and Mexico. The present version of LANaPDB has a total of 13 578 compounds, coming from ten databases of seven Latin American countries. A chemoinformatic characterization of LANaPDB was carried out, which includes the structural classification of the compounds, calculation of six physicochemical properties of pharmaceutical interest, and visualization of the chemical space by employing and comparing two different fingerprints (MACCS keys (166-bit) and Morgan2 (2048-bit)). Furthermore, additional analyses were made, and valuable information not included in the first version of LANaPDB was added, which includes structural diversity, molecular complexity, synthetic feasibility, commercial availability, and reported and predicted biological activity. In addition, the LANaPDB compounds were cross-referenced to two of the largest public chemical compound databases annotated with biological activity: ChEMBL and PubChem.
Collapse
Affiliation(s)
- Alejandro Gómez-García
- DIFACQUIM
Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| | - Daniel A Acuña Jiménez
- CBio3
Laboratory, School of Chemistry, University
of Costa Rica, San Pedro, San José 11501-2060, Costa Rica
| | - William J Zamora
- CBio3
Laboratory, School of Chemistry, University
of Costa Rica, San Pedro, San José 11501-2060, Costa Rica
- Laboratory
of Computational Toxicology and Artificial Intelligence (LaToxCIA),
Biological Testing Laboratory (LEBi), University
of Costa Rica, San Pedro, San José 11501-2060, Costa Rica
- Advanced
Computing Lab (CNCA), National High Technology
Center (CeNAT), Pavas, San José 1174-1200, Costa Rica
| | - Haruna L Barazorda-Ccahuana
- Computational
Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
| | - Miguel Á. Chávez-Fumagalli
- Computational
Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru
| | - Marilia Valli
- School of
Pharmaceutical Sciences of Ribeirao Preto (FCFRP), University of São Paulo (USP), Avenida Professor Doutor Zeferino Vaz, s/n, Ribeirao Preto 14040-903, SP, Brazil
| | - Adriano D Andricopulo
- Laboratory
of Medicinal and Computational Chemistry (LQMC), Centre for Research
and Innovation in Biodiversity and Drug Discovery (CIBFar), São
Carlos Institute of Physics (IFSC), University
of São Paulo (USP), Av. João Dagnone, 1100, São
Carlos 13563-120, SP, Brazil
| | - Vanderlan da S Bolzani
- Nuclei
of Bioassays, Biosynthesis and Ecophysiology of Natural Products (NuBBE),
Department of Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Av. Prof. Francisco Degni, 55, Araraquara 14800-900, SP, Brazil
| | - Dionisio A Olmedo
- Center
for Pharmacognostic Research on Panamanian Flora (CIFLORPAN), College
of Pharmacy, University of Panama, Av. Manuel E. Batista and Jose De
Fabrega, Panama City 3366, Panama
| | - Pablo N Solís
- Center
for Pharmacognostic Research on Panamanian Flora (CIFLORPAN), College
of Pharmacy, University of Panama, Av. Manuel E. Batista and Jose De
Fabrega, Panama City 3366, Panama
| | - Marvin J Núñez
- Natural
Product Research Laboratory, School of Chemistry and Pharmacy, University of El Salvador, Final Ave. Mártires Estudiantes del 30 de
Julio, San Salvador 01101, El Salvador
| | - Johny R Rodríguez Pérez
- GIFAMol
Research Group, School of Chemistry Technology, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
- GIEPRONAL
Research Group, School of Basic Sciences, Technology and Engineering, Universidad Nacional Abierta y a Distancia, Dosquebradas 661001, Colombia
| | - Hoover A Valencia Sánchez
- GIFAMol
Research Group, School of Chemistry Technology, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | - Héctor F Cortés Hernández
- GIFAMol
Research Group, School of Chemistry Technology, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | - Oscar M Mosquera Martinez
- GBPN
Research Group, School of Chemistry Technology, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | - José L Medina-Franco
- DIFACQUIM
Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico
| |
Collapse
|
9
|
Pliatsika D, Blatter C, Riedl R. Targeted protein degradation: current molecular targets, localization, and strategies. Drug Discov Today 2024; 29:104178. [PMID: 39276920 DOI: 10.1016/j.drudis.2024.104178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Targeted protein degradation (TPD) has revolutionized drug discovery by selectively eliminating specific proteins within and outside the cellular context. Over the past two decades, TPD has expanded its focus beyond well-established targets, exploring diverse proteins beyond cancer-related ones. This evolution extends the potential of TPD to various diseases. Notably, TPD can target proteins at demanding locations, such as the extracellular matrix (ECM) and cellular membranes, presenting both opportunities and challenges for future research. In this review, we comprehensively examine the exciting opportunities in the burgeoning field of TPD, highlighting different targets, their cellular environment, and innovative strategies for modern drug discovery.
Collapse
Affiliation(s)
- Dimanthi Pliatsika
- Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences, CH-8820 Wädenswil, Switzerland
| | - Cindy Blatter
- Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences, CH-8820 Wädenswil, Switzerland
| | - Rainer Riedl
- Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences, CH-8820 Wädenswil, Switzerland.
| |
Collapse
|
10
|
de Brevern AG. Special Issue: "Molecular Dynamics Simulations and Structural Analysis of Protein Domains". Int J Mol Sci 2024; 25:10793. [PMID: 39409122 PMCID: PMC11477144 DOI: 10.3390/ijms251910793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
The 3D protein structure is the basis for all their biological functions [...].
Collapse
Affiliation(s)
- Alexandre G. de Brevern
- DSIMB Bioinformatics Team, BIGR, INSERM, Université Paris Cité, F-75015 Paris, France; ; Tel.: +33-1-4449-3000
- DSIMB Bioinformatics Team, BIGR, INSERM, Université de la Réunion, F-97715 Saint Denis, France
| |
Collapse
|
11
|
Zuo WF, Pang Q, Zhu X, Yang QQ, Zhao Q, He G, Han B, Huang W. Heat shock proteins as hallmarks of cancer: insights from molecular mechanisms to therapeutic strategies. J Hematol Oncol 2024; 17:81. [PMID: 39232809 PMCID: PMC11375894 DOI: 10.1186/s13045-024-01601-1] [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: 07/04/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Heat shock proteins are essential molecular chaperones that play crucial roles in stabilizing protein structures, facilitating the repair or degradation of damaged proteins, and maintaining proteostasis and cellular functions. Extensive research has demonstrated that heat shock proteins are highly expressed in cancers and closely associated with tumorigenesis and progression. The "Hallmarks of Cancer" are the core features of cancer biology that collectively define a series of functional characteristics acquired by cells as they transition from a normal state to a state of tumor growth, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabled replicative immortality, the induction of angiogenesis, and the activation of invasion and metastasis. The pivotal roles of heat shock proteins in modulating the hallmarks of cancer through the activation or inhibition of various signaling pathways has been well documented. Therefore, this review provides an overview of the roles of heat shock proteins in vital biological processes from the perspective of the hallmarks of cancer and summarizes the small-molecule inhibitors that target heat shock proteins to regulate various cancer hallmarks. Moreover, we further discuss combination therapy strategies involving heat shock proteins and promising dual-target inhibitors to highlight the potential of targeting heat shock proteins for cancer treatment. In summary, this review highlights how targeting heat shock proteins could regulate the hallmarks of cancer, which will provide valuable information to better elucidate and understand the roles of heat shock proteins in oncology and the mechanisms of cancer occurrence and development and aid in the development of more efficacious and less toxic novel anticancer agents.
Collapse
Affiliation(s)
- Wei-Fang Zuo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiwen Pang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xinyu Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian-Qian Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian Zhao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gu He
- Department of Dermatology and Venereology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
12
|
Sergeeva KV, Tyganov SA, Zaripova KA, Bokov RO, Nikitina LV, Konstantinova TS, Kalamkarov GR, Shenkman BS. Mechanical and signaling responses of unloaded rat soleus muscle to chronically elevated β-myosin activity. Arch Biochem Biophys 2024; 754:109961. [PMID: 38492659 DOI: 10.1016/j.abb.2024.109961] [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: 10/26/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
It has been reported that muscle functional unloading is accompanied by an increase in motoneuronal excitability despite the elimination of afferent input. Thus, we hypothesized that pharmacological potentiation of spontaneous contractile soleus muscle activity during hindlimb unloading could activate anabolic signaling pathways and prevent the loss of muscle mass and strength. To investigate these aspects and underlying molecular mechanisms, we used β-myosin allosteric effector Omecamtiv Mekarbil (OM). We found that OM partially prevented the loss of isometric strength and intrinsic stiffness of the soleus muscle after two weeks of disuse. Notably, OM was able to attenuate the unloading-induced decrease in the rate of muscle protein synthesis (MPS). At the same time, the use of drug neither prevented the reduction in the markers of translational capacity (18S and 28S rRNA) nor activation of the ubiquitin-proteosomal system, which is evidenced by a decrease in the cross-sectional area of fast and slow muscle fibers. These results suggest that chemically-induced increase in low-intensity spontaneous contractions of the soleus muscle during functional unloading creates prerequisites for protein synthesis. At the same time, it should be assumed that the use of OM is advisable with pharmacological drugs that inhibit the expression of ubiquitin ligases.
Collapse
Affiliation(s)
- K V Sergeeva
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia.
| | - S A Tyganov
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - K A Zaripova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - R O Bokov
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - L V Nikitina
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, Russia
| | - T S Konstantinova
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - G R Kalamkarov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - B S Shenkman
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
13
|
Slane EG, Tambrini SJ, Cummings BS. Therapeutic potential of lipin inhibitors for the treatment of cancer. Biochem Pharmacol 2024; 222:116106. [PMID: 38442792 DOI: 10.1016/j.bcp.2024.116106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/28/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Lipins are phosphatidic acid phosphatases (PAP) that catalyze the conversion of phosphatidic acid (PA) to diacylglycerol (DAG). Three lipin isoforms have been identified: lipin-1, -2 and -3. In addition to their PAP activity, lipin-1 and -2 act as transcriptional coactivators and corepressors. Lipins have been intensely studied for their role in regulation of lipid metabolism and adipogenesis; however, lipins are hypothesized to mediate several pathologies, such as those involving metabolic diseases, neuropathy and even cognitive impairment. Recently, an emerging role for lipins have been proposed in cancer. The study of lipins in cancer has been hampered by lack of inhibitors that have selectivity for lipins, that differentiate between lipin family members, or that are suitable for in vivo studies. Such inhibitors have the potential to be extremely useful as both molecular tools and therapeutics. This review describes the expression and function of lipins in various tissues and their roles in several diseases, but with an emphasis on their possible role in cancer. The mechanisms by which lipins mediate cancer cell growth are discussed and the potential usefulness of selective lipin inhibitors is hypothesized. Finally, recent studies reporting the crystallization of lipin-1 are discussed to facilitate rational design of novel lipin inhibitors.
Collapse
Affiliation(s)
- Elizabeth G Slane
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samantha J Tambrini
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Brian S Cummings
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|