1
|
Blockade of NMT1 enzymatic activity inhibits N-myristoylation of VILIP3 protein and suppresses liver cancer progression. Signal Transduct Target Ther 2023; 8:14. [PMID: 36617552 PMCID: PMC9826789 DOI: 10.1038/s41392-022-01248-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/24/2022] [Accepted: 11/01/2022] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. Identification of the underlying mechanism of HCC progression and exploration of new therapeutic drugs are urgently needed. Here, a compound library consisting of 419 FDA-approved drugs was taken to screen potential anticancer drugs. A series of functional assays showed that desloratadine, an antiallergic drug, can repress proliferation in HCC cell lines, cell-derived xenograft (CDX), patient-derived organoid (PDO) and patient-derived xenograft (PDX) models. N-myristoyl transferase 1 (NMT1) was identified as a target protein of desloratadine by drug affinity responsive target stability (DARTS) and surface plasmon resonance (SPR) assays. Upregulation of NMT1 expression enhanced but NMT1 knockdown suppressed tumor growth in vitro and in vivo. Metabolic labeling and mass spectrometry analyses revealed that Visinin-like protein 3 (VILIP3) was a new substrate of NMT1 in protein N-myristoylation modification, and high NMT1 or VILIP3 expression was associated with advanced stages and poor survival in HCC. Mechanistically, desloratadine binds to Asn-246 in NMT1 and inhibits its enzymatic activity, disrupting the NMT1-mediated myristoylation of the VILIP3 protein and subsequent NFκB/Bcl-2 signaling. Conclusively, this study demonstrates that desloratadine may be a novel anticancer drug and that NMT1-mediated myristoylation contributes to HCC progression and is a potential biomarker and therapeutic target in HCC.
Collapse
|
2
|
Wu J, Wang W, Shao X, Lin G, Wang X. Facing the CDK4/6i resistance dilemma in patients with breast cancer, exploration of the resistance mechanism and possible reverse strategy: A narrative review. Medicine (Baltimore) 2022; 101:e32238. [PMID: 36595763 PMCID: PMC9794308 DOI: 10.1097/md.0000000000032238] [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] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is one of the highest rates of malignancy of women, approximate 70% metastatic breast cancer are hormone receptor positive (HR+) and human epidermal growth factor receptor 2 negative (HER2-). Hormone therapy is the primary strategy of HR+/HER2- metastatic breast cancer. With the permission of cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i), progress free survival and overall survival were significantly licensed. However, inevitable outcome of CDK4/6i resistance has become the main reason that restricts the clinical benefit of patients. In recent years, the research on dealing with drug resistance has become a hot topic, a large number of molecular mechanisms have been focused, and a lot of experiments have been carried out at the preclinical level. This review summarizes the current knowledge of CDK4/6i resistance mechanism, systematically expounds the signaling pathways and targets leading to CDK4/6i resistance, analyzes different ways and mechanisms, and provides theoretical guidance for the clinical reversal of endocrine therapy resistance.
Collapse
Affiliation(s)
- Jiayi Wu
- Department of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei Wang
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiying Shao
- Department of Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China
- *Correspondence: Xiaojia Wang, Department of Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China (e-mail: ) and Xiying Shao, Department of Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China (e-mail: )
| | - Guang Lin
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaojia Wang
- Department of Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China
- *Correspondence: Xiaojia Wang, Department of Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China (e-mail: ) and Xiying Shao, Department of Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China (e-mail: )
| |
Collapse
|
3
|
Sharma D, Kumar N, Mehrotra T, Pervaiz N, Agrawal L, Tripathi S, Jha A, Poullikkas T, Kumar R, Ledwani L. In vitro and in silico molecular docking studies of Rheum emodi-derived diamagnetic SnO 2 nanoparticles and their cytotoxic effects against breast cancer. NEW J CHEM 2021. [DOI: 10.1039/d0nj04670a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Green-route-bioengineered nanoparticles have received significant attention for diagnosis and treatment of cancer in the medical technology era due to their non-toxic nature, cost-friendliness, and energy efficiency.
Collapse
Affiliation(s)
| | - Naveen Kumar
- Department of Physics, Shri Guru Gobind Singh College
- Chandigarh
- India
| | - Tarang Mehrotra
- College of Professional Studies
- Northeastern University
- Boston
- USA
| | | | - Lokesh Agrawal
- Universidad Integral del Caribe y América Latina, Kaminda Cas Grandi #79
- Curaçao
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University
- Okinawa 904-0412
- Japan
| | - Shalini Tripathi
- Department of Material Science
- University of Connecticut
- Storrs
- USA
| | - Abhishek Jha
- Department of Life Sciences
- School of Sciences
- Indira Gandhi National Open University
- India
| | - Thanasis Poullikkas
- Human Biology, School of Integrative and Global Majors, University of Tsukuba
- 1-1-1 Tennodai
- Japan
- Department of Experimental Pathology, Faculty of Medicine, University of Tsukuba
- 2-1-1 Tennodai
| | | | | |
Collapse
|
4
|
N-myristoyltransferase-1 is necessary for lysosomal degradation and mTORC1 activation in cancer cells. Sci Rep 2020; 10:11952. [PMID: 32686708 PMCID: PMC7371688 DOI: 10.1038/s41598-020-68615-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022] Open
Abstract
N-myristoyltransferase-1 (NMT1) catalyzes protein myristoylation, a lipid modification that is elevated in cancer cells. NMT1 sustains proliferation and/or survival of cancer cells through mechanisms that are not completely understood. We used genetic and pharmacological inhibition of NMT1 to further dissect the role of this enzyme in cancer, and found an unexpected essential role for NMT1 at promoting lysosomal metabolic functions. Lysosomes mediate enzymatic degradation of vesicle cargo, and also serve as functional platforms for mTORC1 activation. We show that NMT1 is required for both lysosomal functions in cancer cells. Inhibition of NMT1 impaired lysosomal degradation leading to autophagy flux blockade, and simultaneously caused the dissociation of mTOR from the surface of lysosomes leading to decreased mTORC1 activation. The regulation of lysosomal metabolic functions by NMT1 was largely mediated through the lysosomal adaptor LAMTOR1. Accordingly, genetic targeting of LAMTOR1 recapitulated most of the lysosomal defects of targeting NMT1, including defective lysosomal degradation. Pharmacological inhibition of NMT1 reduced tumor growth, and tumors from treated animals had increased apoptosis and displayed markers of lysosomal dysfunction. Our findings suggest that compounds targeting NMT1 may have therapeutic benefit in cancer by preventing mTORC1 activation and simultaneously blocking lysosomal degradation, leading to cancer cell death.
Collapse
|
5
|
Kosciuk T, Lin H. N-Myristoyltransferase as a Glycine and Lysine Myristoyltransferase in Cancer, Immunity, and Infections. ACS Chem Biol 2020; 15:1747-1758. [PMID: 32453941 DOI: 10.1021/acschembio.0c00314] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein myristoylation, the addition of a 14-carbon saturated acyl group, is an abundant modification implicated in biological events as diverse as development, immunity, oncogenesis, and infections. N-Myristoyltransferase (NMT) is the enzyme that catalyzes this modification. Many elegant studies have established the rules guiding the catalysis including substrate amino acid sequence requirements with the indispensable N-terminal glycine, and a co-translational mode of action. Recent advances in technology such as the development of fatty acid analogs, small molecule inhibitors, and new proteomic strategies, allowed a deeper insight into the NMT activity and function. Here we focus on discussing recent work demonstrating that NMT is also a lysine myristoyltransferase, the enzyme's regulation by a previously unnoticed solvent channel, and the mechanism of NMT regulation by protein-protein interactions. We also summarize recent findings on NMT's role in cancer, immunity, and infections and the advances in pharmacological targeting of myristoylation. Our analyses highlight opportunities for further understanding and discoveries.
Collapse
Affiliation(s)
- Tatsiana Kosciuk
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, United States
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
6
|
Portet S. A primer on model selection using the Akaike Information Criterion. Infect Dis Model 2020; 5:111-128. [PMID: 31956740 PMCID: PMC6962709 DOI: 10.1016/j.idm.2019.12.010] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 12/27/2019] [Indexed: 11/24/2022] Open
Abstract
A powerful investigative tool in biology is to consider not a single mathematical model but a collection of models designed to explore different working hypotheses and select the best model in that collection. In these lecture notes, the usual workflow of the use of mathematical models to investigate a biological problem is described and the use of a collection of model is motivated. Models depend on parameters that must be estimated using observations; and when a collection of models is considered, the best model has then to be identified based on available observations. Hence, model calibration and selection, which are intrinsically linked, are essential steps of the workflow. Here, some procedures for model calibration and a criterion, the Akaike Information Criterion, of model selection based on experimental data are described. Rough derivation, practical technique of computation and use of this criterion are detailed.
Collapse
Affiliation(s)
- Stéphanie Portet
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| |
Collapse
|
7
|
Presti D, Quaquarini E. The PI3K/AKT/mTOR and CDK4/6 Pathways in Endocrine Resistant HR+/HER2- Metastatic Breast Cancer: Biological Mechanisms and New Treatments. Cancers (Basel) 2019; 11:E1242. [PMID: 31450618 PMCID: PMC6770492 DOI: 10.3390/cancers11091242] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/17/2019] [Accepted: 08/20/2019] [Indexed: 12/30/2022] Open
Abstract
Endocrine-based treatments are the normal standard-of-care in women with hormone receptor-positive/Human Epidermal growth factor Receptor 2-negative metastatic breast cancer. Despite the well-known efficacy of these drugs as first-line therapies, about 50% of women develop endocrine resistance and disease progression. The treatment of these patients has represented one of the most important research fields in the last few years, with several multicenter phase II/III trials published or still ongoing. Novel therapies, such as cyclin-dependent kinase (CDK)4/6 and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) inhibitors, have significantly changed the prognosis of patients progressing to a previous endocrine treatment, allowing a great benefit in terms of progression-free survival and, in some cases, of overall survival. However, identifying response predictors is essential for the rational use of these drugs to avoid unnecessary toxicity and costs, and to ensure the optimal therapeutic sequence is used. In this review, we analyze the PI3K/AKT/mTOR and CDK4/6 pathways and their roles in endocrine resistant metastatic breast cancer. We then focus on the new treatments developed and the roles of these drugs in overcoming endocrine resistance, describing the latest clinical trials that led to the approval of the drugs in clinical practice.
Collapse
Affiliation(s)
- Daniele Presti
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
- Medical Oncology Unit, IRCCS ICS Maugeri SpA SB, 27100 Pavia, Italy
| | - Erica Quaquarini
- Medical Oncology Unit, IRCCS ICS Maugeri SpA SB, 27100 Pavia, Italy.
- Experimental Medicine, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|