1
|
Sastourné-Haletou R, Marynberg S, Pereira A, Su F, Chen M, Valet G, Sindikubwabo F, Cañeque T, Müller S, Colombeau L, Solier S, Gaillet C, Guianvarc'h D, Biot C, Karoyan P, Gueroui Z, Arimondo P, Klausen M, Vauzeilles B, Cossy J, Fontecave M, Gasser G, Policar C, Gautier A, Johannes L, Rodriguez R. PSL Chemical Biology Symposia: The Increasing Impact of Chemistry in Life Sciences. Chembiochem 2025:e2500231. [PMID: 40195606 DOI: 10.1002/cbic.202500231] [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] [Received: 03/18/2025] [Indexed: 04/09/2025]
Abstract
This symposium is the 6th Paris Sciences & Lettres (PSL) Chemical Biology meeting (2015, 2016, 2019, 2023, 2024, 2025) being held at Institut Curie. This initiative originally started in 2013 at Institut de Chimie des Substances Naturelles (ICSN) in Gif-sur-Yvette and was mostly focused on organic synthesis. It was then exported at Institut Curie to cover a larger scope, before becoming the official French Chemical Biology meeting. This year, around 200 participants had the opportunity to meet world leaders in chemistry and biology who described their latest innovations and future trends covering topics as diverse as prebiotic chemistry, activity-based protein profiling, high-resolution cell imaging, nanotechnologies, bio-orthogonal chemistry, metal ion signaling, ferroptosis, and biocatalysis.
Collapse
Affiliation(s)
- Romain Sastourné-Haletou
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Sacha Marynberg
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Arthur Pereira
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Fubao Su
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Mengnuo Chen
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Gaspard Valet
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Fabien Sindikubwabo
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Tatiana Cañeque
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Sebastian Müller
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Ludovic Colombeau
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Stéphanie Solier
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Christine Gaillet
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | | | - Christophe Biot
- UGSF, Université de Lille, UMR 8576 CNRS, 59655, Villeneuve d'Ascq, France
| | - Philippe Karoyan
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Zoher Gueroui
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Paola Arimondo
- Chimie Biologique Épigénétique, Institut Pasteur, UMR 3523 CNRS, 75724, Paris, France
| | - Maxime Klausen
- ICB, Chimie ParisTech, UMR 8060 CNRS, 75005, Paris, France
| | - Boris Vauzeilles
- ICSN, Université Paris-Saclay, UPR 2301 CNRS, 91190, Gif-sur-Yvette, France
| | | | - Marc Fontecave
- LCPB, Collège de France, UMR 8229 CNRS, 75005, Paris, France
| | - Gilles Gasser
- ICB, Chimie ParisTech, UMR 8060 CNRS, 75005, Paris, France
| | - Clotilde Policar
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Arnaud Gautier
- CPCV, École Normale Supérieure, Sorbonne Université, UMR 8228 CNRS, 75005, Paris, France
| | - Ludger Johannes
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| | - Raphaël Rodriguez
- Chemical Biology of Cancer, Institut Curie, PSL Université Paris, UMR 3666 CNRS, U1339 INSERM, 75005, Paris, France
| |
Collapse
|
2
|
Xiang Y, Tanwar V, Singh P, Follette LL, Narayan V, Kapahi P. Early menarche and childbirth accelerate aging-related outcomes and age-related diseases: Evidence for antagonistic pleiotropy in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.09.23.24314197. [PMID: 39398990 PMCID: PMC11469407 DOI: 10.1101/2024.09.23.24314197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Aging can be understood as a consequence of the declining force of natural selection with age. Consistent with this, the antagonistic pleiotropy theory of aging proposes that aging arises from trade-offs that favor early growth and reproduction. However, evidence supporting antagonistic pleiotropy in humans remains limited. Using Mendelian Randomization (MR), we demonstrated that later ages of menarche or first childbirth were genetically associated with longer parental lifespan, decreased frailty index, slower epigenetic aging, later menopause, and reduced facial aging. Moreover, later menarche or first childbirth were also genetically associated with a lower risk of several age-related diseases, including late-onset Alzheimer's disease (LOAD), type 2 diabetes, heart disease, essential hypertension, and chronic obstructive pulmonary disease (COPD). We validated the associations between the age of menarche, childbirth, and the number of childbirths with several age-related outcomes in the UK Biobank by conducting regression analysis of nearly 200,000 subjects. Our results demonstrated that menarche before the age 11 and childbirth before 21 significantly accelerated the risk of several diseases, and almost doubled the risk for diabetes, heart failure, and quadrupled the risk of obesity, supporting the antagonistic pleiotropy theory. We identified 126 significant single nucleotide polymorphisms (SNPs) that influenced age-related outcomes, some of which were involved in known longevity pathways, including IGF1, growth hormone, AMPK, and mTOR signaling. Our study also identified higher BMI as a mediating factor in causing the increased risk of certain diseases, such as type 2 diabetes and heart failure, in women with early menarche or early pregnancy, emphasizing the importance of the thrifty gene hypothesis in explaining in part the mechanisms behind antagonistic pleiotropy. Our study highlights the complex relationship between genetic legacies and modern diseases, emphasizing the need for gender-sensitive healthcare strategies that consider the unique connections between female reproductive health and aging.
Collapse
Affiliation(s)
- Yifan Xiang
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Vineeta Tanwar
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Parminder Singh
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | | | - Vikram Narayan
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
| | - Pankaj Kapahi
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA 94143
| |
Collapse
|
3
|
Orobator ON, Mertens RT, Obisesan OA, Awuah SG. Energy and endoplasmic reticulum stress induction by gold(III) dithiocarbamate and 2-deoxyglucose synergistically trigger cell death in breast cancer. J Biol Chem 2024:107949. [PMID: 39481597 DOI: 10.1016/j.jbc.2024.107949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
The elusiveness of triple-negative breast cancer from targeted therapy has redirected focus towards exploiting the metabolic shortcomings of these highly metastatic subtypes of breast cancer. Cueing from the metabolic heterogeneity of TNBC and the exposition of the dual dependence of some TNBCs on OXPHOS and glycolysis for ATP, we herein report the efficacy of cotreatment of TNBCs with an OXPHOS inhibitor, 2a and 2DG, a potent glycolysis inhibitor. 2a-2DG cotreatment inhibited TNBC cell proliferation with IC50 of ∼5 to 36 times lower than that of 2a alone and over 5000 times lower than IC50 of 2DG alone. 2a-2DG cotreatment suppressed mitochondrial ATP production and significantly induced AMPK activation. Mechanistic studies revealed the distinct yet synergistic contributions of 2a and 2DG to the antiproliferative effect of the cotreatment. While 2a induced apoptotic cell death, 2DG sensitized TNBCs to the antiproliferative effects of 2a via endoplasmic reticulum stress induction. Strikingly, the combination of 2a-2DG ablated SUM159 tumors in an orthotopic xenograft mouse model. This study highlights the synergistic effect of a gold-based complex with 2DG and the potential benefit of multi-metabolic pathways targeting as an effective therapeutic strategy against TNBCs.
Collapse
Affiliation(s)
- Owamagbe N Orobator
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Oluwatosin A Obisesan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States; Center for Pharmaceutical Research and Innovation, Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA; Center for Bioelectronics and Nanomedicine, University of Kentucky, Lexington KY, 40506, USA.
| |
Collapse
|
4
|
Witwit H, Betancourt CA, Cubitt B, Khafaji R, Kowalski H, Jackson N, Ye C, Martinez-Sobrido L, de la Torre JC. Cellular N-Myristoyl Transferases Are Required for Mammarenavirus Multiplication. Viruses 2024; 16:1362. [PMID: 39339839 PMCID: PMC11436053 DOI: 10.3390/v16091362] [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: 08/02/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
The mammarenavirus matrix Z protein plays critical roles in virus assembly and cell egress. Meanwhile, heterotrimer complexes of a stable signal peptide (SSP) together with glycoprotein subunits GP1 and GP2, generated via co-and post-translational processing of the surface glycoprotein precursor GPC, form the spikes that decorate the virion surface and mediate virus cell entry via receptor-mediated endocytosis. The Z protein and the SSP undergo N-terminal myristoylation by host cell N-myristoyltransferases (NMT1 and NMT2), and G2A mutations that prevent myristoylation of Z or SSP have been shown to affect the Z-mediated virus budding and GP2-mediated fusion activity that is required to complete the virus cell entry process. In the present work, we present evidence that the validated on-target specific pan-NMT inhibitor DDD85646 exerts a potent antiviral activity against the prototypic mammarenavirus lymphocytic choriomeningitis virus (LCMV) that correlates with reduced Z budding activity and GP2-mediated fusion activity as well as with proteasome-mediated degradation of the Z protein. The potent anti-mammarenaviral activity of DDD85646 was also observed with the hemorrhagic-fever-causing Junin (JUNV) and Lassa (LASV) mammarenaviruses. Our results support the exploration of NMT inhibition as a broad-spectrum antiviral against human pathogenic mammarenaviruses.
Collapse
Affiliation(s)
- Haydar Witwit
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (H.W.); (C.A.B.); (B.C.); (R.K.)
| | - Carlos Alberto Betancourt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (H.W.); (C.A.B.); (B.C.); (R.K.)
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (H.W.); (C.A.B.); (B.C.); (R.K.)
| | - Roaa Khafaji
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (H.W.); (C.A.B.); (B.C.); (R.K.)
| | - Heinrich Kowalski
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria;
| | - Nathaniel Jackson
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (N.J.); (L.M.-S.)
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (N.J.); (L.M.-S.)
| | | | - Juan C. de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (H.W.); (C.A.B.); (B.C.); (R.K.)
| |
Collapse
|
5
|
Witwit H, Betancourt C, Cubitt B, Khafaji R, Kowalski H, Jackson N, Ye C, Martinez-Sobrido L, de la Torre JC. Cellular N-myristoyl transferases Are Required for Mammarenavirus Multiplication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606235. [PMID: 39211253 PMCID: PMC11361045 DOI: 10.1101/2024.08.01.606235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The mammarenavirus matrix Z protein plays critical roles in virus assembly and cell egress, whereas heterotrimer complexes of a stable signal peptide (SSP) together with glycoprotein subunits GP1 and GP2, generated via co-and post-translational processing of the surface glycoprotein precursor GPC, form the spikes that decorate the virion surface and mediate virus cell entry via receptor-mediated endocytosis. The Z protein and SSP undergo N-terminal myristoylation by host cell N-myristoyltransferases (NMT1 and NMT2), and G2A mutations that prevent myristoylation of Z or SSP have been shown to affect Z mediated virus budding and GP2 mediated fusion activity required to complete the virus cell entry process. In the present work, we present evidence that the validated on-target specific pan NMT inhibitor DDD85464 exerts a potent antiviral activity against the prototypic mammarenavirus lymphocytic choriomeningitis virus (LCMV) that correlated with reduced Z budding activity and GP2 mediated fusion activity, as well as proteasome mediated degradation of the Z protein. The potent anti-mammarenaviral activity of DDD85646 was also observed with the hemorrhagic fever causing mammarenaviruses Junin (JUNV) and Lassa (LASV) viruses. Our results support exploration of NMT inhibition as a broad-spectrum antiviral against human pathogenic mammarenaviruses.
Collapse
|
6
|
Tan X, He Y, Yu P, Deng Y, Xie Z, Guo J, Hou Q, Li P, Lin X, Ouyang S, Ma W, Xie Y, Guo Z, Chen D, Zhang Z, Zhu Y, Huang F, Zhao Z, Zhang C, Guo Z, Chen X, Peng T, Li L, Xie W. The dual role of FSP1 in programmed cell death: resisting ferroptosis in the cell membrane and promoting necroptosis in the nucleus of THP-1 cells. Mol Med 2024; 30:102. [PMID: 39009982 PMCID: PMC11247902 DOI: 10.1186/s10020-024-00861-4] [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: 02/02/2024] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Acute monocytic leukemia-M5 (AML-M5) remains a challenging disease due to its high morbidity and poor prognosis. In addition to the evidence mentioned earlier, several studies have shown that programmed cell death (PCD) serves a critical function in treatment of AML-M5. However, the role and relationship between ferroptosis and necroptosis in AML-M5 remains unclear. METHODS THP-1 cells were mainly treated with Erastin and IMP-366. The changes of ferroptosis and necroptosis levels were detected by CCK-8, western blot, quantitative real-time PCR, and electron microscopy. Flow cytometry was applied to detect the ROS and lipid ROS levels. MDA, 4-HNE, GSH and GSSG were assessed by ELISA kits. Intracellular distribution of FSP1 was studied by immunofluorescent staining and western blot. RESULTS The addition of the myristoylation inhibitor IMP-366 to erastin-treated acute monocytic leukemia cell line THP-1 cell not only resulted in greater susceptibility to ferroptosis characterized by lipid peroxidation, glutathione (GSH) depletion and mitochondrial shrinkage, as the FSP1 position on membrane was inhibited, but also increased p-RIPK1 and p-MLKL protein expression, as well as a decrease in caspase-8 expression, and triggered the characteristic necroptosis phenomena, including cytoplasmic translucency, mitochondrial swelling, membranous fractures by FSP1 migration into the nucleus via binding importin α2. It is interesting to note that ferroptosis inhibitor fer-1 reversed necroptosis. CONCLUSION We demonstrated that inhibition of myristoylation by IMP-366 is capable of switching ferroptosis and ferroptosis-dependent necroptosis in THP-1 cells. In these findings, FSP1-mediated ferroptosis and necroptosis are described as alternative mechanisms of PCD of THP-1 cells, providing potential therapeutic strategies and targets for AML-M5.
Collapse
Affiliation(s)
- Xiaoqian Tan
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Yinling He
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Panpan Yu
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yunong Deng
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiami Guo
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qin Hou
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wentao Ma
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zilong Guo
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Dandan Chen
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zhixia Zhang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Yunyu Zhu
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Fei Huang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Cen Zhang
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Zhirong Guo
- Class of Clinical Medicine, University of South China, Hengyang, 421001, Hunan, China
| | - Xi Chen
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Liang Li
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Department of Physiology, Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
7
|
Geroyska S, Mejia I, Chan AA, Navarrete M, Pandey V, Kharpatin S, Noguti J, Wang F, Srole D, Chou TF, Wohlschlegel J, Nemeth E, Damoiseaux R, Shackelford DB, Lee DJ, Díaz B. N-Myristoytransferase Inhibition Causes Mitochondrial Iron Overload and Parthanatos in TIM17A-Dependent Aggressive Lung Carcinoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1815-1833. [PMID: 38949950 PMCID: PMC11270646 DOI: 10.1158/2767-9764.crc-23-0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 05/09/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Myristoylation is a type of protein acylation by which the fatty acid myristate is added to the N-terminus of target proteins, a process mediated by N-myristoyltransferases (NMT). Myristoylation is emerging as a promising cancer therapeutic target; however, the molecular determinants of sensitivity to NMT inhibition or the mechanism by which it induces cancer cell death are not completely understood. We report that NMTs are a novel therapeutic target in lung carcinoma cells with LKB1 and/or KEAP1 mutations in a KRAS-mutant background. Inhibition of myristoylation decreases cell viability in vitro and tumor growth in vivo. Inhibition of myristoylation causes mitochondrial ferrous iron overload, oxidative stress, elevated protein poly (ADP)-ribosylation, and death by parthanatos. Furthermore, NMT inhibitors sensitized lung carcinoma cells to platinum-based chemotherapy. Unexpectedly, the mitochondrial transporter translocase of inner mitochondrial membrane 17 homolog A (TIM17A) is a critical target of myristoylation inhibitors in these cells. TIM17A silencing recapitulated the effects of NMT inhibition at inducing mitochondrial ferrous iron overload and parthanatos. Furthermore, sensitivity of lung carcinoma cells to myristoylation inhibition correlated with their dependency on TIM17A. This study reveals the unexpected connection between protein myristoylation, the mitochondrial import machinery, and iron homeostasis. It also uncovers myristoylation inhibitors as novel inducers of parthanatos in cancer, and the novel axis NMT-TIM17A as a potential therapeutic target in highly aggressive lung carcinomas. SIGNIFICANCE KRAS-mutant lung carcinomas with LKB1 and/or KEAP1 co-mutations have intrinsic therapeutic resistance. We show that these tumors are sensitive to NMT inhibitors, which slow tumor growth in vivo and sensitize cells to platinum-based chemotherapy in vitro. Inhibition of myristoylation causes death by parthanatos and thus has the potential to kill apoptosis and ferroptosis-resistant cancer cells. Our findings warrant investigation of NMT as a therapeutic target in highly aggressive lung carcinomas.
Collapse
Affiliation(s)
- Sofia Geroyska
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Hematology and Oncology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Isabel Mejia
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Hematology and Oncology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Alfred A. Chan
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Marian Navarrete
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Vijaya Pandey
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Samuel Kharpatin
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Juliana Noguti
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Feng Wang
- Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| | - Daniel Srole
- UCLA Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Tsui-Fen Chou
- Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| | - James Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Elizabeta Nemeth
- UCLA Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California.
- California NanoSystems Institute at UCLA, Los Angeles, California.
- Department for Bioengineering, Samueli School of Engineering, UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| | - David B. Shackelford
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| | - Delphine J. Lee
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| | - Begoña Díaz
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Hematology and Oncology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| |
Collapse
|
8
|
Beauchamp E, Gamma JM, Cromwell CR, Moussa EW, Pain R, Kostiuk MA, Acevedo-Morantes C, Iyer A, Yap M, Vincent KM, Postovit LM, Julien O, Hubbard BP, Mackey JR, Berthiaume LG. Multiomics analysis identifies oxidative phosphorylation as a cancer vulnerability arising from myristoylation inhibition. J Transl Med 2024; 22:431. [PMID: 38715059 PMCID: PMC11075276 DOI: 10.1186/s12967-024-05150-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/31/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND In humans, two ubiquitously expressed N-myristoyltransferases, NMT1 and NMT2, catalyze myristate transfer to proteins to facilitate membrane targeting and signaling. We investigated the expression of NMTs in numerous cancers and found that NMT2 levels are dysregulated by epigenetic suppression, particularly so in hematologic malignancies. This suggests that pharmacological inhibition of the remaining NMT1 could allow for the selective killing of these cells, sparing normal cells with both NMTs. METHODS AND RESULTS Transcriptomic analysis of 1200 NMT inhibitor (NMTI)-treated cancer cell lines revealed that NMTI sensitivity relates not only to NMT2 loss or NMT1 dependency, but also correlates with a myristoylation inhibition sensitivity signature comprising 54 genes (MISS-54) enriched in hematologic cancers as well as testis, brain, lung, ovary, and colon cancers. Because non-myristoylated proteins are degraded by a glycine-specific N-degron, differential proteomics revealed the major impact of abrogating NMT1 genetically using CRISPR/Cas9 in cancer cells was surprisingly to reduce mitochondrial respiratory complex I proteins rather than cell signaling proteins, some of which were also reduced, albeit to a lesser extent. Cancer cell treatments with the first-in-class NMTI PCLX-001 (zelenirstat), which is undergoing human phase 1/2a trials in advanced lymphoma and solid tumors, recapitulated these effects. The most downregulated myristoylated mitochondrial protein was NDUFAF4, a complex I assembly factor. Knockout of NDUFAF4 or in vitro cell treatment with zelenirstat resulted in loss of complex I, oxidative phosphorylation and respiration, which impacted metabolomes. CONCLUSIONS Targeting of both, oxidative phosphorylation and cell signaling partly explains the lethal effects of zelenirstat in select cancer types. While the prognostic value of the sensitivity score MISS-54 remains to be validated in patients, our findings continue to warrant the clinical development of zelenirstat as cancer treatment.
Collapse
Affiliation(s)
| | - Jay M Gamma
- Department of Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Christopher R Cromwell
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Eman W Moussa
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rony Pain
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Morris A Kostiuk
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Claudia Acevedo-Morantes
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aishwarya Iyer
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Megan Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Krista M Vincent
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lynne M Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Olivier Julien
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Basil P Hubbard
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - Luc G Berthiaume
- Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada.
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
9
|
Tate EW, Soday L, de la Lastra AL, Wang M, Lin H. Protein lipidation in cancer: mechanisms, dysregulation and emerging drug targets. Nat Rev Cancer 2024; 24:240-260. [PMID: 38424304 DOI: 10.1038/s41568-024-00666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 03/02/2024]
Abstract
Protein lipidation describes a diverse class of post-translational modifications (PTMs) that is regulated by over 40 enzymes, targeting more than 1,000 substrates at over 3,000 sites. Lipidated proteins include more than 150 oncoproteins, including mediators of cancer initiation, progression and immunity, receptor kinases, transcription factors, G protein-coupled receptors and extracellular signalling proteins. Lipidation regulates the physical interactions of its protein substrates with cell membranes, regulating protein signalling and trafficking, and has a key role in metabolism and immunity. Targeting protein lipidation, therefore, offers a unique approach to modulate otherwise undruggable oncoproteins; however, the full spectrum of opportunities to target the dysregulation of these PTMs in cancer remains to be explored. This is attributable in part to the technological challenges of identifying the targets and the roles of protein lipidation. The early stage of drug discovery for many enzymes in the pathway contrasts with efforts for drugging similarly common PTMs such as phosphorylation and acetylation, which are routinely studied and targeted in relevant cancer contexts. Here, we review recent advances in identifying targetable protein lipidation pathways in cancer, the current state-of-the-art in drug discovery, and the status of ongoing clinical trials, which have the potential to deliver novel oncology therapeutics targeting protein lipidation.
Collapse
Affiliation(s)
- Edward W Tate
- Department of Chemistry, Imperial College London, London, UK.
- Francis Crick Institute, London, UK.
| | - Lior Soday
- Department of Chemistry, Imperial College London, London, UK
| | | | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
| | - Hening Lin
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
10
|
Andrade C, Sousa BKDP, Sigurdardóttir S, Bourgard C, Borba J, Clementino L, Salazar-Alvarez LC, Groustra S, Zigweid R, Khim M, Staker B, Costa F, Eriksson L, Sunnerhagen P. Selective Bias Virtual Screening for Discovery of Promising Antimalarial Candidates targeting Plasmodium N-Myristoyltransferase. RESEARCH SQUARE 2024:rs.3.rs-3963523. [PMID: 38463971 PMCID: PMC10925453 DOI: 10.21203/rs.3.rs-3963523/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Malaria remains a significant public health challenge, with Plasmodium vivax being the species responsible for the most prevalent form of the disease. Given the limited therapeutic options available, the search for new antimalarials against P. vivax is urgent. This study aims to identify new inhibitors for P. vivax N-myristoyltransferase (PvNMT), an essential drug target against malaria. Through a validated virtual screening campaign, we prioritized 23 candidates for further testing. In the yeast NMT system, seven compounds exhibit a potential inhibitor phenotype. In vitro antimalarial phenotypic assays confirmed the activity of four candidates while demonstrating an absence of cytotoxicity. Enzymatic assays reveal LabMol-394 as the most promising inhibitor, displaying selectivity against the parasite and a strong correlation within the yeast system. Furthermore, molecular dynamics simulations shed some light into its binding mode. This study constitutes a substantial contribution to the exploration of a selective quinoline scaffold and provides valuable insights into the development of new antimalarial candidates.
Collapse
|
11
|
McHugh D, Sun B, Gutierrez-Muñoz C, Hernández-González F, Mellone M, Guiho R, Duran I, Pombo J, Pietrocola F, Birch J, Kallemeijn WW, Khadayate S, Dharmalingam G, Vernia S, Tate EW, Martínez-Barbera JP, Withers DJ, Thomas GJ, Serrano M, Gil J. COPI vesicle formation and N-myristoylation are targetable vulnerabilities of senescent cells. Nat Cell Biol 2023; 25:1804-1820. [PMID: 38012402 PMCID: PMC10709147 DOI: 10.1038/s41556-023-01287-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/12/2023] [Indexed: 11/29/2023]
Abstract
Drugs that selectively kill senescent cells (senolytics) improve the outcomes of cancer, fibrosis and age-related diseases. Despite their potential, our knowledge of the molecular pathways that affect the survival of senescent cells is limited. To discover senolytic targets, we performed RNAi screens and identified coatomer complex I (COPI) vesicle formation as a liability of senescent cells. Genetic or pharmacological inhibition of COPI results in Golgi dispersal, dysfunctional autophagy, and unfolded protein response-dependent apoptosis of senescent cells, and knockdown of COPI subunits improves the outcomes of cancer and fibrosis in mouse models. Drugs targeting COPI have poor pharmacological properties, but we find that N-myristoyltransferase inhibitors (NMTi) phenocopy COPI inhibition and are potent senolytics. NMTi selectively eliminated senescent cells and improved outcomes in models of cancer and non-alcoholic steatohepatitis. Our results suggest that senescent cells rely on a hyperactive secretory apparatus and that inhibiting trafficking kills senescent cells with the potential to treat various senescence-associated diseases.
Collapse
Affiliation(s)
- Domhnall McHugh
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Bin Sun
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Carmen Gutierrez-Muñoz
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Fernanda Hernández-González
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Pulmonology, ICR, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Massimiliano Mellone
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- AstraZeneca, Immuno-Oncology Discovery, Oncology R&D, Cambridge, UK
| | - Romain Guiho
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Imanol Duran
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Joaquim Pombo
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Federico Pietrocola
- Karolinska Institute, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Jodie Birch
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Wouter W Kallemeijn
- Department of Chemistry, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Sanjay Khadayate
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Gopuraja Dharmalingam
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Santiago Vernia
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Juan Pedro Martínez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dominic J Withers
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Gareth J Thomas
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Altos Labs, Cambridge Institute of Science, Granta Park, UK
| | - Jesús Gil
- MRC Laboratory of Medical Sciences (LMS), London, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
12
|
Abstract
N-myristoyltransferase 1 (NMT1) is an indispensable eukaryotic enzyme that catalyses the transfer of myristoyl groups to the amino acid terminal residues of numerous proteins. This catalytic process is required for the growth and development of many eukaryotes and viruses. Elevated expression and activity of NMT1 is observed to varying degrees in a variety of tumour types (e.g. colon, lung and breast tumours). Furthermore, an elevated level of NMT1 in tumours is associated with poor survival. Therefore, a relationship exists between NMT1 and tumours. In this review, we discuss the underlying mechanisms by which NMT1 is associated with tumour development from the perspective of oncogene signalling, involvement in cellular metabolism, and endoplasmic reticulum stress. Several NMT inhibitors used in cancer treatment are introduced. The review will provide some directions for future research.Key MessagesElevated expression and activity of NMT1 is observed to varying degrees in a variety of tumour types which creates the possibility of targeting NMT1 in tumours.NMT1-mediated myristoylation plays a pivotal role in cancer cell metabolism and may be particularly relevant to cancer metastasis and drug resistance. These insights can be used to direct potential therapeutic avenues for NMT1 inhibitors.
Collapse
Affiliation(s)
- Hong Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Xu
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Thoracic OncologyShanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Thoracic OncologyShanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Medical Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongxia Qiao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Chang YH. Impact of Protein N α-Modifications on Cellular Functions and Human Health. Life (Basel) 2023; 13:1613. [PMID: 37511988 PMCID: PMC10381334 DOI: 10.3390/life13071613] [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: 06/21/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Most human proteins are modified by enzymes that act on the α-amino group of a newly synthesized polypeptide. Methionine aminopeptidases can remove the initiator methionine and expose the second amino acid for further modification by enzymes responsible for myristoylation, acetylation, methylation, or other chemical reactions. Specific acetyltransferases can also modify the initiator methionine and sometimes the acetylated methionine can be removed, followed by further modifications. These modifications at the protein N-termini play critical roles in cellular protein localization, protein-protein interaction, protein-DNA interaction, and protein stability. Consequently, the dysregulation of these modifications could significantly change the development and progression status of certain human diseases. The focus of this review is to highlight recent progress in our understanding of the roles of these modifications in regulating protein functions and how these enzymes have been used as potential novel therapeutic targets for various human diseases.
Collapse
Affiliation(s)
- Yie-Hwa Chang
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University Medical School, Saint Louis, MO 63104, USA
| |
Collapse
|
14
|
Spassov DS, Atanasova M, Doytchinova I. Inhibitor Trapping in N-Myristoyltransferases as a Mechanism for Drug Potency. Int J Mol Sci 2023; 24:11610. [PMID: 37511367 PMCID: PMC10380619 DOI: 10.3390/ijms241411610] [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] [Received: 06/27/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Predicting inhibitor potency is critical in drug design and development, yet it has remained one of computational biology's biggest unresolved challenges. Here, we show that in the case of the N-myristoyltransferase (NMT), this problem could be traced to the mechanisms by which the NMT enzyme is inhibited. NMT adopts open or closed conformations necessary for orchestrating the different steps of the catalytic process. The results indicate that the potency of the NMT inhibitors is determined by their ability to stabilize the enzyme conformation in the closed state, and that in this state, the small molecules themselves are trapped and locked inside the structure of the enzyme, creating a significant barrier for their dissociation. By using molecular dynamics simulations, we demonstrate that the conformational stabilization of the protein molecule in its closed form is highly correlated with the ligands activity and can be used to predict their potency. Hence, predicting inhibitor potency in silico might depend on modeling the conformational changes of the protein molecule upon binding of the ligand rather than estimating the changes in free binding energy that arise from their interaction.
Collapse
Affiliation(s)
- Danislav S Spassov
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Mariyana Atanasova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Irini Doytchinova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| |
Collapse
|
15
|
SPI1 Mediates N-Myristoyltransferase 1 to Advance Gastric Cancer Progression via PI3K/AKT/mTOR Pathway. Can J Gastroenterol Hepatol 2023; 2023:2021515. [PMID: 36967718 PMCID: PMC10038735 DOI: 10.1155/2023/2021515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 03/19/2023] Open
Abstract
Gastric cancer (GC) is a common digestive tract malignancy worldwide. N-myristoyltransferase 1 (NMT1) has been implicated in many cancers, but its association with gastric cancer remains to be clarified. Thus, this paper elucidated the role of NMT1 in GC. The NMT1 expression level in GC and normal tissue samples as well as the relationship between NMT1 high or low expression and overall survival in GC was analyzed via GEPIA. GC cells were transfected with NMT1 or SPI1 overexpression plasmid and short hairpin RNA against NMT1 (shNMT1) or shSPI1. NMT1, SPI1, p-PI3K, PI3K, p-AKT, AKT, p-mTOR, and mTOR levels were detected through qRT-PCR and western blot. MTT, wound healing, and transwell assays were applied to test cell viability, migration, and invasion. The binding relationship of SPI1 and NMT1 was determined through a dual-luciferase reporter assay and chromatin immunoprecipitation. NMT1 was upregulated in GC, the high level of which connected with a poor prognosis. Overexpressed NMT1 elevated viability, migration rate, and invasion rate of GC cells, whereas NMT1 knockdown leads to the opposite results. Besides, SPI1 could bind to NMT1. Overexpressed NMT1 reversed the effects of shSPI1 on decreasing viability, migration, invasion, p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR in GC cells, and NMT1 knockdown reversed the effects of SPI1 overexpression on increasing viability, migration, invasion, p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR. SPI1 upregulated NMT1 to facilitate the malignant behaviors of GC cells through the PI3K/AKT/mTOR pathway.
Collapse
|
16
|
Lemarié FL, Sanders SS, Nguyen Y, Martin DDO, Hayden MR. Full-length huntingtin is palmitoylated at multiple sites and post-translationally myristoylated following caspase-cleavage. Front Physiol 2023; 14:1086112. [PMID: 36711022 PMCID: PMC9880554 DOI: 10.3389/fphys.2023.1086112] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Introduction: Huntington disease is an autosomal dominant neurodegenerative disorder which is caused by a CAG repeat expansion in the HTT gene that codes for an elongated polyglutamine tract in the huntingtin (HTT) protein. Huntingtin is subjected to multiple post-translational modifications which regulate its cellular functions and degradation. We have previously identified a palmitoylation site at cysteine 214 (C214), catalyzed by the enzymes ZDHHC17 and ZDHHC13. Reduced palmitoylation level of mutant huntingtin is linked to toxicity and loss of function. Moreover, we have described N-terminal myristoylation by the N-myristoyltransferases of a short fragment of huntingtin (HTT553-586) at glycine 553 (G553) following proteolysis at aspartate 552 (D552). Results: Here, we show that huntingtin is palmitoylated at numerous cysteines: C105, C433, C3134 and C3144. In addition, we confirm that full-length huntingtin is cleaved at D552 and post-translationally myristoylated at G553. Importantly, blocking caspase cleavage at the critical and pathogenic aspartate 586 (D586) significantly increases posttranslational myristoylation of huntingtin. In turn, myristoylation of huntingtin promotes the co-interaction between C-terminal and N-terminal huntingtin fragments, which is also protective. Discussion: This suggests that the protective effect of inhibiting caspase-cleavage at D586 may be mediated through post-translational myristoylation of huntingtin at G553.
Collapse
|
17
|
Spassov DS, Atanasova M, Doytchinova I. A role of salt bridges in mediating drug potency: A lesson from the N-myristoyltransferase inhibitors. Front Mol Biosci 2023; 9:1066029. [PMID: 36703920 PMCID: PMC9871453 DOI: 10.3389/fmolb.2022.1066029] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
The salt bridge is the strongest non-covalent interaction in nature and is known to participate in protein folding, protein-protein interactions, and molecular recognition. However, the role of salt bridges in the context of drug design has remained not well understood. Here, we report that a common feature in the mechanism of inhibition of the N-myristoyltransferases (NMT), promising targets for the treatment of protozoan infections and cancer, is the formation of a salt bridge between a positively charged chemical group of the small molecule and the negatively charged C-terminus of the enzyme. Substituting the inhibitor positively charged amine group with a neutral methylene group prevents the formation of the salt bridge and leads to a dramatic activity loss. Molecular dynamics simulations have revealed that salt bridges stabilize the NMT-ligand complexes by functioning as molecular clips that stabilize the conformation of the protein structure. As such, the creation of salt bridges between the ligands and their protein targets may find an application as a valuable tool in rational drug design.
Collapse
|
18
|
Mohammadinejad A, Mohajeri T, Aleyaghoob G, Heidarian F, Kazemi Oskuee R. Ellagic acid as a potent anticancer drug: A comprehensive review on in vitro, in vivo, in silico, and drug delivery studies. Biotechnol Appl Biochem 2022; 69:2323-2356. [PMID: 34846078 DOI: 10.1002/bab.2288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/10/2021] [Indexed: 12/27/2022]
Abstract
Ellagic acid as a polyphenol or micronutrient, which can be naturally found in different vegetables and fruits, has gained considerable attention for cancer therapy due to considerable biological activities and different molecular targets. Ellagic acid with low hydrolysis and lipophilic and hydrophobic nature is not able to be absorbed in circulation. So, accumulation inside the intestinal epithelial cells or metabolization to other urolithins leads to the limitation of direct evaluation of EA effects in clinical studies. This review focuses on the studies which supported anticancer activity of pure or fruit-extracted ellagic acid through in vitro, in vivo, in silico, and drug delivery methods. The results demonstrate ellagic acid modulates the expression of various genes incorporated in the cancer-related process of apoptosis and proliferation, inflammation related-gens, and oxidative-related genes. Moreover, the ellagic acid formulation in carriers composed of lipid, silica, chitosan, iron- bovine serum albumin nanoparticles obviously enhanced the stable release and confident delivery with minimum loss. Also, in silico analysis proved that ellagic acid was able to be placed at a position of cocrystal ADP, in the deep cavity of the protein target, and tightly interact with binding pocket residues leading to suppression of substrate availability of protein and its activation inhibition.
Collapse
Affiliation(s)
- Arash Mohammadinejad
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Taraneh Mohajeri
- Department of Obstetrics & Gynecology, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Ghazaleh Aleyaghoob
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Heidarian
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Novel Hits for N-Myristoyltransferase Inhibition Discovered by Docking-Based Screening. Molecules 2022; 27:molecules27175478. [PMID: 36080246 PMCID: PMC9457982 DOI: 10.3390/molecules27175478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/26/2022] Open
Abstract
N-myristoyltransferase (NMT) inhibitors that were initially developed for treatment of parasitic protozoan infections, including sleeping sickness, malaria, and leismaniasis, have also shown great promise as treatment for oncological diseases. The successful transition of NMT inhibitors, which are currently at preclinical to early clinical stages, toward clinical approval and utilization may depend on the development and design of a diverse set of drug molecules with particular selectivity or pharmacological properties. In our study, we report that a common feature in the inhibitory mechanism of NMT is the formation of a salt bridge between a positively charged chemical group of the small molecule and the negatively charged C-terminus of an enzyme. Based on this observation, we designed a virtual screening protocol to identify novel ligands that mimic this mode of interaction. By screening over 1.1 million structures downloaded from the ZINC database, several hits were identified that displayed NMT inhibitory activity. The stability of the inhibitor-NMT complexes was evaluated by molecular dynamics simulations. The ligands from the stable complexes were tested in vitro and some of them appear to be promising leads for further optimization.
Collapse
|
20
|
Xu Y, Shi Z, Bao L. An expanding repertoire of protein acylations. Mol Cell Proteomics 2022; 21:100193. [PMID: 34999219 PMCID: PMC8933697 DOI: 10.1016/j.mcpro.2022.100193] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/22/2021] [Accepted: 01/04/2022] [Indexed: 01/03/2023] Open
Abstract
Protein post-translational modifications play key roles in multiple cellular processes by allowing rapid reprogramming of individual protein functions. Acylation, one of the most important post-translational modifications, is involved in different physiological activities including cell differentiation and energy metabolism. In recent years, the progression in technologies, especially the antibodies against acylation and the highly sensitive and effective mass spectrometry–based proteomics, as well as optimized functional studies, greatly deepen our understanding of protein acylation. In this review, we give a general overview of the 12 main protein acylations (formylation, acetylation, propionylation, butyrylation, malonylation, succinylation, glutarylation, palmitoylation, myristoylation, benzoylation, crotonylation, and 2-hydroxyisobutyrylation), including their substrates (histones and nonhistone proteins), regulatory enzymes (writers, readers, and erasers), biological functions (transcriptional regulation, metabolic regulation, subcellular targeting, protein–membrane interactions, protein stability, and folding), and related diseases (cancer, diabetes, heart disease, neurodegenerative disease, and viral infection), to present a complete picture of protein acylations and highlight their functional significance in future research. Provide a general overview of the 12 main protein acylations. Acylation of viral proteins promotes viral integration and infection. Hyperacylation of histone has antitumous and neuroprotective effects. MS is widely used in the identification of acylation but has its challenges.
Collapse
Affiliation(s)
- Yuxuan Xu
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research center for Cancer, 300060, Tianjin, China
| | - Zhenyu Shi
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research center for Cancer, 300060, Tianjin, China
| | - Li Bao
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research center for Cancer, 300060, Tianjin, China.
| |
Collapse
|
21
|
Giglione C, Meinnel T. Mapping the myristoylome through a complete understanding of protein myristoylation biochemistry. Prog Lipid Res 2021; 85:101139. [PMID: 34793862 DOI: 10.1016/j.plipres.2021.101139] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/22/2022]
Abstract
Protein myristoylation is a C14 fatty acid modification found in all living organisms. Myristoylation tags either the N-terminal alpha groups of cysteine or glycine residues through amide bonds or lysine and cysteine side chains directly or indirectly via glycerol thioester and ester linkages. Before transfer to proteins, myristate must be activated into myristoyl coenzyme A in eukaryotes or, in bacteria, to derivatives like phosphatidylethanolamine. Myristate originates through de novo biosynthesis (e.g., plants), from external uptake (e.g., human tissues), or from mixed origins (e.g., unicellular organisms). Myristate usually serves as a molecular anchor, allowing tagged proteins to be targeted to membranes and travel across endomembrane networks in eukaryotes. In this review, we describe and discuss the metabolic origins of protein-bound myristate. We review strategies for in vivo protein labeling that take advantage of click-chemistry with reactive analogs, and we discuss new approaches to the proteome-wide discovery of myristate-containing proteins. The machineries of myristoylation are described, along with how protein targets can be generated directly from translating precursors or from processed proteins. Few myristoylation catalysts are currently described, with only N-myristoyltransferase described to date in eukaryotes. Finally, we describe how viruses and bacteria hijack and exploit myristoylation for their pathogenicity.
Collapse
Affiliation(s)
- Carmela Giglione
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France.
| | - Thierry Meinnel
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France.
| |
Collapse
|
22
|
Kallemeijn WW, Lanyon-Hogg T, Panyain N, Goya Grocin A, Ciepla P, Morales-Sanfrutos J, Tate EW. Proteome-wide analysis of protein lipidation using chemical probes: in-gel fluorescence visualization, identification and quantification of N-myristoylation, N- and S-acylation, O-cholesterylation, S-farnesylation and S-geranylgeranylation. Nat Protoc 2021; 16:5083-5122. [PMID: 34707257 DOI: 10.1038/s41596-021-00601-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/05/2021] [Indexed: 02/08/2023]
Abstract
Protein lipidation is one of the most widespread post-translational modifications (PTMs) found in nature, regulating protein function, structure and subcellular localization. Lipid transferases and their substrate proteins are also attracting increasing interest as drug targets because of their dysregulation in many disease states. However, the inherent hydrophobicity and potential dynamic nature of lipid modifications makes them notoriously challenging to detect by many analytical methods. Chemical proteomics provides a powerful approach to identify and quantify these diverse protein modifications by combining bespoke chemical tools for lipidated protein enrichment with quantitative mass spectrometry-based proteomics. Here, we report a robust and proteome-wide approach for the exploration of five major classes of protein lipidation in living cells, through the use of specific chemical probes for each lipid PTM. In-cell labeling of lipidated proteins is achieved by the metabolic incorporation of a lipid probe that mimics the specific natural lipid, concomitantly wielding an alkyne as a bio-orthogonal labeling tag. After incorporation, the chemically tagged proteins can be coupled to multifunctional 'capture reagents' by using click chemistry, allowing in-gel fluorescence visualization or enrichment via affinity handles for quantitative chemical proteomics based on label-free quantification (LFQ) or tandem mass-tag (TMT) approaches. In this protocol, we describe the application of lipid probes for N-myristoylation, N- and S-acylation, O-cholesterylation, S-farnesylation and S-geranylgeranylation in multiple cell lines to illustrate both the workflow and data obtained in these experiments. We provide detailed workflows for method optimization, sample preparation for chemical proteomics and data processing. A properly trained researcher (e.g., technician, graduate student or postdoc) can complete all steps from optimizing metabolic labeling to data processing within 3 weeks. This protocol enables sensitive and quantitative analysis of lipidated proteins at a proteome-wide scale at native expression levels, which is critical to understanding the role of lipid PTMs in health and disease.
Collapse
Affiliation(s)
- Wouter W Kallemeijn
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Thomas Lanyon-Hogg
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Nattawadee Panyain
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Global Health Institute, Faculty of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Andrea Goya Grocin
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Paulina Ciepla
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Institute of Chemical Sciences and Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Julia Morales-Sanfrutos
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
23
|
Schlott AC, Knuepfer E, Green JL, Hobson P, Borg AJ, Morales-Sanfrutos J, Perrin AJ, Maclachlan C, Collinson LM, Snijders AP, Tate EW, Holder AA. Inhibition of protein N-myristoylation blocks Plasmodium falciparum intraerythrocytic development, egress and invasion. PLoS Biol 2021; 19:e3001408. [PMID: 34695132 PMCID: PMC8544853 DOI: 10.1371/journal.pbio.3001408] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022] Open
Abstract
We have combined chemical biology and genetic modification approaches to investigate the importance of protein myristoylation in the human malaria parasite, Plasmodium falciparum. Parasite treatment during schizogony in the last 10 to 15 hours of the erythrocytic cycle with IMP-1002, an inhibitor of N-myristoyl transferase (NMT), led to a significant blockade in parasite egress from the infected erythrocyte. Two rhoptry proteins were mislocalized in the cell, suggesting that rhoptry function is disrupted. We identified 16 NMT substrates for which myristoylation was significantly reduced by NMT inhibitor (NMTi) treatment, and, of these, 6 proteins were substantially reduced in abundance. In a viability screen, we showed that for 4 of these proteins replacement of the N-terminal glycine with alanine to prevent myristoylation had a substantial effect on parasite fitness. In detailed studies of one NMT substrate, glideosome-associated protein 45 (GAP45), loss of myristoylation had no impact on protein location or glideosome assembly, in contrast to the disruption caused by GAP45 gene deletion, but GAP45 myristoylation was essential for erythrocyte invasion. Therefore, there are at least 3 mechanisms by which inhibition of NMT can disrupt parasite development and growth: early in parasite development, leading to the inhibition of schizogony and formation of “pseudoschizonts,” which has been described previously; at the end of schizogony, with disruption of rhoptry formation, merozoite development and egress from the infected erythrocyte; and at invasion, when impairment of motor complex function prevents invasion of new erythrocytes. These results underline the importance of P. falciparum NMT as a drug target because of the pleiotropic effect of its inhibition. Understanding the essential factors needed for malaria parasite development could help us find new therapeutic targets. This study reveals that N-myristoylation is a posttranslational modification of proteins essential for the parasites’ growth and their invasion of red blood cells.
Collapse
Affiliation(s)
- Anja C. Schlott
- Malaria Parasitology Laboratory, Francis Crick Institute, London, United Kingdom
- Molecular Sciences Research Hub, Imperial College, London, United Kingdom
| | - Ellen Knuepfer
- Malaria Parasitology Laboratory, Francis Crick Institute, London, United Kingdom
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, United Kingdom
| | - Judith L. Green
- Malaria Parasitology Laboratory, Francis Crick Institute, London, United Kingdom
| | - Philip Hobson
- Flow Cytometry Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | - Aaron J. Borg
- Mass Spectrometry Proteomics Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | | | - Abigail J. Perrin
- Malaria Biochemistry Laboratory, Francis Crick Institute, London, United Kingdom
| | - Catherine Maclachlan
- Electron Microscopy Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | - Ambrosius P. Snijders
- Mass Spectrometry Proteomics Science Technology Platform, Francis Crick Institute, London, United Kingdom
| | - Edward W. Tate
- Molecular Sciences Research Hub, Imperial College, London, United Kingdom
- Francis Crick Institute, London, United Kingdom
- * E-mail: (EWT); (AAH)
| | - Anthony A. Holder
- Malaria Parasitology Laboratory, Francis Crick Institute, London, United Kingdom
- * E-mail: (EWT); (AAH)
| |
Collapse
|
24
|
Tang H, Cui M, Han M. Fatty acids impact sarcomere integrity through myristoylation and ER homeostasis. Cell Rep 2021; 36:109539. [PMID: 34407398 PMCID: PMC8404530 DOI: 10.1016/j.celrep.2021.109539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 06/04/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Decreased ability to maintain tissue integrity is critically involved in aging and degenerative diseases. Fatty acid (FA) metabolism has a profound impact on animal development and tissue maintenance, but our understanding of the underlying mechanisms is limited. We investigated whether and how FA abundance affects muscle integrity using Caenorhabditis elegans. We show that reducing the overall FA level by blocking FA biosynthesis or inhibiting protein myristoylation leads to disorganization of sarcomere structure and adult-onset paralysis. Further analysis indicates that myristoylation of two ARF guanosine triphosphatases (GTPases) critically mediates the effect of FA deficiency on sarcomere integrity through inducing endoplasmic reticulum (ER) stress and ER unfolded protein response (UPRER), which in turn leads to reduction of the level of sarcomere component PINCH and myosin disorganization. We thus present a mechanism that links FA signal, protein myristoylation, and ER homeostasis with muscle integrity, which provides valuable insights into the regulatory role of nutrients and ER homeostasis in muscle maintenance.
Collapse
Affiliation(s)
- Hongyun Tang
- Department of MCDB, University of Colorado Boulder, Boulder, CO 80309, USA; Key Laboratory of Growth Regulation and Transformation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang Province, China; Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Mingxue Cui
- Department of MCDB, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Min Han
- Department of MCDB, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
25
|
Suazo KF, Park KY, Distefano MD. A Not-So-Ancient Grease History: Click Chemistry and Protein Lipid Modifications. Chem Rev 2021; 121:7178-7248. [PMID: 33821625 PMCID: PMC8820976 DOI: 10.1021/acs.chemrev.0c01108] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein lipid modification involves the attachment of hydrophobic groups to proteins via ester, thioester, amide, or thioether linkages. In this review, the specific click chemical reactions that have been employed to study protein lipid modification and their use for specific labeling applications are first described. This is followed by an introduction to the different types of protein lipid modifications that occur in biology. Next, the roles of click chemistry in elucidating specific biological features including the identification of lipid-modified proteins, studies of their regulation, and their role in diseases are presented. A description of the use of protein-lipid modifying enzymes for specific labeling applications including protein immobilization, fluorescent labeling, nanostructure assembly, and the construction of protein-drug conjugates is presented next. Concluding remarks and future directions are presented in the final section.
Collapse
Affiliation(s)
- Kiall F. Suazo
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Keun-Young Park
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Mark D. Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| |
Collapse
|
26
|
Zhu G, Wang F, Li H, Zhang X, Wu Q, Liu Y, Qian M, Guo S, Yang Y, Xue X, Sun F, Qiao Y, Pan Q. N-Myristoylation by NMT1 Is POTEE-Dependent to Stimulate Liver Tumorigenesis via Differentially Regulating Ubiquitination of Targets. Front Oncol 2021; 11:681366. [PMID: 34136404 PMCID: PMC8201403 DOI: 10.3389/fonc.2021.681366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/03/2021] [Indexed: 01/15/2023] Open
Abstract
Background A tremendous amount of studies have suggested that post-translational modifications (PTMs) play pivotal roles during tumorigenesis. Compared to other PTMs, lipid modification is less studied. Recently, N-myristoylation, one type of lipid modification, has been paid attention to the field of cancer. However, whether and how N-myristoylation exerts its roles in liver tumorigenesis still remains unclear. Methods Parallel reaction monitoring (PRM) was conducted to evaluate the expression of protein modification enzymes in paired tissues. Liver conditionally knocking NMT1 out mice model was used to assess the critical roles of N-myristoylation during liver tumorigenesis. Proteomics isobaric tags for relative and absolute quantification (iTraq) was performed to identify proteins that changed while NMT1 was knocked down. The click chemistry assay was used to evaluate the N-myristoylation levels of proteins. Results Here, N-myristolyation and its enzyme NMT1, but not NMT2, were found to be critical in liver cancer. Two categories of proteins, i.e., N-myristolyation down-regulated proteins (NDP, including LXN, RPL29, and FAU) and N-myristolyation up-regulated proteins (NUP, including AHSG, ALB, and TF), were revealed negatively and positively regulated by NMT1, respectively. Both NDP and NUP could be N-myristolyated by NMT1 indispensable of POTEE. However, N-myristolyation decreased and increased stability of NDP and NUP, respectively. Mechanistically, NDP-specific binding protein RPL7A facilitated HIST1H4H, which has ubiquitin E3 ligase function, to ubiquitinate NDP. By contrast, NUP-specific binding protein HBB prevented NUP from ubiquitination by HIST1H4H. Notably, function of RPL7A and HBB was all NMT1-dependent. Moreover, NDP suppressed while NUP stimulated transformative phenotypes. Clinically, higher levels of NMT1 and NUP with lower levels of NDP had worse prognostic outcome. Conclusion Collectively, N-myristolyation by NMT1 suppresses anti-tumorigenic NDP, whereas it stimulates pro-tumorigenic NUP by interfering their ubiquitination to finally result in a pro-tumorigenic outcome in liver cancer. Targeting N-myristolyation and NMT1 might be helpful to treat liver cancer.
Collapse
Affiliation(s)
- Guoqing Zhu
- Department of Clinical Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Feng Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Haojie Li
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Wu
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Ya Liu
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Mingping Qian
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Susu Guo
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yueyue Yang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangfei Xue
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Yongxia Qiao
- School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiuhui Pan
- Department of Clinical Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
27
|
(Carboxymethyl-stevioside)-coated magnetic dots for enhanced magnetic hyperthermia and improved glioblastoma treatment. Colloids Surf B Biointerfaces 2021; 205:111870. [PMID: 34034224 DOI: 10.1016/j.colsurfb.2021.111870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022]
Abstract
The use of different types of biomaterials as surfactant moities has a defined role in magnetic hyperthermia-mediated cancer therapy (MHCT). In this work, we present carboxymethyl-stevioside (CMS)-modified magnetic dots (MDs) as efficient magnetic hyperthermia agents for glioma therapy. The synthesized MDs with CMS biosurfactant coating exhibited significant water stability that resulted in a remarkable specific absorption rate of 209.25 W/g on application of alternating magnetic field of strength 359 kHz and 188 Oe. The MDs further demonstrated significant anti-migratory and anti-invasive effect on glioma C6 cells by inhibiting the gene expression of matrix metalloproteinases-2 and -9. The effect of immediate and long term hyperthermia treatment was then evaluated after repetitive exposure to hyperthermia, in terms of glioma cell viability, the effect of treatment on cell morphology, the cell cycle distribution and oxidative stress generation. The results obtained suggest the promising potential of CMS-modified nano-heaters for excellent magnetic hyperthermia-mediated glioma therapy.
Collapse
|
28
|
Xu J, Taubert S. Beyond Proteostasis: Lipid Metabolism as a New Player in ER Homeostasis. Metabolites 2021; 11:52. [PMID: 33466824 PMCID: PMC7830277 DOI: 10.3390/metabo11010052] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Biological membranes are not only essential barriers that separate cellular and subcellular structures, but also perform other critical functions such as the initiation and propagation of intra- and intercellular signals. Each membrane-delineated organelle has a tightly regulated and custom-made membrane lipid composition that is critical for its normal function. The endoplasmic reticulum (ER) consists of a dynamic membrane network that is required for the synthesis and modification of proteins and lipids. The accumulation of unfolded proteins in the ER lumen activates an adaptive stress response known as the unfolded protein response (UPR-ER). Interestingly, recent findings show that lipid perturbation is also a direct activator of the UPR-ER, independent of protein misfolding. Here, we review proteostasis-independent UPR-ER activation in the genetically tractable model organism Caenorhabditis elegans. We review the current knowledge on the membrane lipid composition of the ER, its impact on organelle function and UPR-ER activation, and its potential role in human metabolic diseases. Further, we summarize the bi-directional interplay between lipid metabolism and the UPR-ER. We discuss recent progress identifying the different respective mechanisms by which disturbed proteostasis and lipid bilayer stress activate the UPR-ER. Finally, we consider how genetic and metabolic disturbances may disrupt ER homeostasis and activate the UPR and discuss how using -omics-type analyses will lead to more comprehensive insights into these processes.
Collapse
Affiliation(s)
- Jiaming Xu
- Graduate Program in Cell and Developmental Biology, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Healthy Starts Theme, British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Stefan Taubert
- Graduate Program in Cell and Developmental Biology, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Healthy Starts Theme, British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
29
|
Zhang Q, Zhou W, Yu S, Ju Y, To SKY, Wong AST, Jiao Y, Poon TCW, Tam KY, Lee LTO. Metabolic reprogramming of ovarian cancer involves ACSL1-mediated metastasis stimulation through upregulated protein myristoylation. Oncogene 2021; 40:97-111. [PMID: 33082557 DOI: 10.1038/s41388-020-01516-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 11/09/2022]
Abstract
As a result of the hostile microenvironment, metabolic alterations are required to enable the malignant growth of cancer cells. To understand metabolic reprogramming during metastasis, we conducted shotgun proteomic analysis of highly metastatic (HM) and non-metastatic (NM) ovarian cancer cells. The results suggest that the genes involved in fatty-acid (FA) metabolism are upregulated, with consequent increases of phospholipids with relatively short FA chains (myristic acid, MA) in HM cells. Among the upregulated proteins, ACSL1 expression could convert the lipid profile of NM cells to that similar of HM cells and make them highly aggressive. Importantly, we demonstrated that ACSL1 activates the AMP-activated protein kinase and Src pathways via protein myristoylation and finally enhances FA beta oxidation. Patient samples and tissue microarray data also suggested that omentum metastatic tumours have higher ACSL1 expression than primary tumours and a strong association with poor clinical outcome. Overall, our data reveal that ACSL1 enhances cancer metastasis by regulating FA metabolism and myristoylation.
Collapse
Affiliation(s)
- Qingyu Zhang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Shan Yu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yaojun Ju
- Proteomics, Metabolomics and Drug Development Core, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Sally Kit Yan To
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Alice Sze Tsai Wong
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yufei Jiao
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Terence Chuen Wai Poon
- Proteomics, Metabolomics and Drug Development Core, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kin Yip Tam
- Proteomics, Metabolomics and Drug Development Core, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Leo Tsz On Lee
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
- Centre of Reproduction, Development, and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
30
|
Beauchamp E, Yap MC, Iyer A, Perinpanayagam MA, Gamma JM, Vincent KM, Lakshmanan M, Raju A, Tergaonkar V, Tan SY, Lim ST, Dong WF, Postovit LM, Read KD, Gray DW, Wyatt PG, Mackey JR, Berthiaume LG. Targeting N-myristoylation for therapy of B-cell lymphomas. Nat Commun 2020; 11:5348. [PMID: 33093447 PMCID: PMC7582192 DOI: 10.1038/s41467-020-18998-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 09/18/2020] [Indexed: 01/05/2023] Open
Abstract
Myristoylation, the N-terminal modification of proteins with the fatty acid myristate, is critical for membrane targeting and cell signaling. Because cancer cells often have increased N-myristoyltransferase (NMT) expression, NMTs were proposed as anti-cancer targets. To systematically investigate this, we performed robotic cancer cell line screens and discovered a marked sensitivity of hematological cancer cell lines, including B-cell lymphomas, to the potent pan-NMT inhibitor PCLX-001. PCLX-001 treatment impacts the global myristoylation of lymphoma cell proteins and inhibits early B-cell receptor (BCR) signaling events critical for survival. In addition to abrogating myristoylation of Src family kinases, PCLX-001 also promotes their degradation and, unexpectedly, that of numerous non-myristoylated BCR effectors including c-Myc, NFκB and P-ERK, leading to cancer cell death in vitro and in xenograft models. Because some treated lymphoma patients experience relapse and die, targeting B-cell lymphomas with a NMT inhibitor potentially provides an additional much needed treatment option for lymphoma.
Collapse
Affiliation(s)
- Erwan Beauchamp
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada.,Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada
| | - Megan C Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada.,Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada
| | - Aishwarya Iyer
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada
| | - Maneka A Perinpanayagam
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada.,Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada
| | - Jay M Gamma
- Departments of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada
| | - Krista M Vincent
- Departments of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada
| | - Manikandan Lakshmanan
- Mouse Models of Human Cancer Unit, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, 138673, Singapore
| | - Anandhkumar Raju
- Advanced Molecular Pathology Lab, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore, 138673.,Department of Pathology, National University of Singapore, Singapore, Singapore
| | - Vinay Tergaonkar
- Advanced Molecular Pathology Lab, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore, 138673.,Department of Pathology, National University of Singapore, Singapore, Singapore
| | - Soo Yong Tan
- Advanced Molecular Pathology Lab, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore, 138673.,Department of Pathology, National University of Singapore, Singapore, Singapore
| | - Soon Thye Lim
- Department of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Outram Road, Singapore, 169610, Singapore
| | - Wei-Feng Dong
- Departments of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada
| | - Lynne M Postovit
- Departments of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada
| | - Kevin D Read
- Drug Discovery Unit, School of Life Sciences, University of Dundee, James Black Centre, Dow Street, Dundee, DD1 5EH, UK
| | - David W Gray
- Drug Discovery Unit, School of Life Sciences, University of Dundee, James Black Centre, Dow Street, Dundee, DD1 5EH, UK
| | - Paul G Wyatt
- Drug Discovery Unit, School of Life Sciences, University of Dundee, James Black Centre, Dow Street, Dundee, DD1 5EH, UK
| | - John R Mackey
- Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada.,Departments of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada
| | - Luc G Berthiaume
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2H7, AB, Canada. .,Pacylex Pharmaceuticals Inc., Edmonton, AB, Canada.
| |
Collapse
|
31
|
Yuan M, Song ZH, Ying MD, Zhu H, He QJ, Yang B, Cao J. N-myristoylation: from cell biology to translational medicine. Acta Pharmacol Sin 2020; 41:1005-1015. [PMID: 32203082 PMCID: PMC7468318 DOI: 10.1038/s41401-020-0388-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Various lipids and lipid metabolites are bound to and modify the proteins in eukaryotic cells, which are known as ‘protein lipidation’. There are four major types of the protein lipidation, i.e. myristoylation, palmitoylation, prenylation, and glycosylphosphatidylinositol anchor. N-myristoylation refers to the attachment of 14-carbon fatty acid myristates to the N-terminal glycine of proteins by N-myristoyltransferases (NMT) and affects their physiology such as plasma targeting, subcellular tracking and localization, thereby influencing the function of proteins. With more novel pathogenic N-myristoylated proteins are identified, the N-myristoylation will attract great attentions in various human diseases including infectious diseases, parasitic diseases, and cancers. In this review, we summarize the current understanding of N-myristoylation in physiological processes and discuss the hitherto implication of crosstalk between N-myristoylation and other protein modification. Furthermore, we mention several well-studied NMT inhibitors mainly in infectious diseases and cancers and generalize the relation of NMT and cancer progression by browsing the clinic database. This review also aims to highlight the further investigation into the dynamic crosstalk of N-myristoylation in physiological processes as well as the potential application of protein N-myristoylation in translational medicine.
Collapse
|
32
|
Bell AS, Yu Z, Hutton JA, Wright MH, Brannigan JA, Paape D, Roberts SM, Sutherell CL, Ritzefeld M, Wilkinson AJ, Smith DF, Leatherbarrow RJ, Tate EW. Novel Thienopyrimidine Inhibitors of Leishmania N-Myristoyltransferase with On-Target Activity in Intracellular Amastigotes. J Med Chem 2020; 63:7740-7765. [PMID: 32575985 PMCID: PMC7383931 DOI: 10.1021/acs.jmedchem.0c00570] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
![]()
The
leishmaniases, caused by Leishmania species
of protozoan parasites, are neglected tropical diseases with millions
of cases worldwide. Current therapeutic approaches are limited by
toxicity, resistance, and cost. N-Myristoyltransferase
(NMT), an enzyme ubiquitous and essential in all eukaryotes, has been
validated via genetic and pharmacological methods as a promising anti-leishmanial
target. Here we describe a comprehensive structure–activity
relationship (SAR) study of a thienopyrimidine series previously identified
in a high-throughput screen against Leishmania NMT,
across 68 compounds in enzyme- and cell-based assay formats. Using
a chemical tagging target engagement biomarker assay, we identify
the first inhibitor in this series with on-target NMT activity in
leishmania parasites. Furthermore, crystal structure analyses of 12
derivatives in complex with Leishmania major NMT revealed key factors important for future structure-guided optimization
delivering IMP-105 (43), a compound with modest activity
against Leishmania donovani intracellular
amastigotes and excellent selectivity (>660-fold) for Leishmania NMT over human NMTs.
Collapse
Affiliation(s)
- Andrew S Bell
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Zhiyong Yu
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Jennie A Hutton
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Megan H Wright
- School of Chemistry, University of Leeds, Leeds, U.K. LS2 9JT
| | - James A Brannigan
- Structural Biology Laboratory, York Biomedical Research Institute, Department of Chemistry, University of York, York, U.K. YO10 5DD
| | - Daniel Paape
- Centre for Immunology and Infection, York Biomedical Research Institute, Department of Biology, University of York, York, U.K. YO10 5NG
| | - Shirley M Roberts
- Structural Biology Laboratory, York Biomedical Research Institute, Department of Chemistry, University of York, York, U.K. YO10 5DD
| | - Charlotte L Sutherell
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Markus Ritzefeld
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Anthony J Wilkinson
- Structural Biology Laboratory, York Biomedical Research Institute, Department of Chemistry, University of York, York, U.K. YO10 5DD
| | - Deborah F Smith
- Centre for Immunology and Infection, York Biomedical Research Institute, Department of Biology, University of York, York, U.K. YO10 5NG
| | - Robin J Leatherbarrow
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, U.K. W12 0BZ
| |
Collapse
|
33
|
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: 18] [Impact Index Per Article: 3.6] [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
|
34
|
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: 2.6] [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
|
35
|
Fedoryshchak RO, Ocasio CA, Strutton B, Mattocks J, Corran AJ, Tate EW. Wheat pathogen Zymoseptoria tritici N-myristoyltransferase inhibitors: on-target antifungal activity and an unusual metabolic defense mechanism. RSC Chem Biol 2020; 1:68-78. [PMID: 34458749 PMCID: PMC8341946 DOI: 10.1039/d0cb00020e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Zymoseptoria tritici is the causative agent of Septoria tritici blotch (STB), which costs billions of dollars annually to major wheat-producing countries in terms of both fungicide use and crop loss. Agricultural pathogenic fungi have acquired resistance to most commercially available fungicide classes, and the rate of discovery and development of new fungicides has stalled, demanding new approaches and insights. Here we investigate a potential mechanism of targeting an important wheat pathogen Z. tritici via inhibition of N-myristoyltransferase (NMT). We characterize Z. tritici NMT biochemically for the first time, profile the in vivo Z. tritici myristoylated proteome and identify and validate the first Z. tritici NMT inhibitors. Proteomic investigation of the downstream effects of NMT inhibition identified an unusual and novel mechanism of defense against chemical toxicity in Z. tritici through the application of comparative bioinformatics to deconvolute function from the previously largely unannotated Z. tritici proteome. Research into novel fungicidal modes-of-action is essential to satisfy an urgent unmet need for novel fungicide targets, and we anticipate that this study will serve as a useful proteomics and bioinformatics resource for researchers studying Z. tritici.
Collapse
Affiliation(s)
- Roman O Fedoryshchak
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub Wood Lane London W12 0BZ UK
- The Francis Crick Institute 1 Midland Rd London NW1 1AT UK
| | - Cory A Ocasio
- The Francis Crick Institute 1 Midland Rd London NW1 1AT UK
| | | | - Jo Mattocks
- Syngenta AG, Jealott's Hill Research Centre Bracknell UK
| | | | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub Wood Lane London W12 0BZ UK
- The Francis Crick Institute 1 Midland Rd London NW1 1AT UK
| |
Collapse
|
36
|
Meinnel T, Dian C, Giglione C. Myristoylation, an Ancient Protein Modification Mirroring Eukaryogenesis and Evolution. Trends Biochem Sci 2020; 45:619-632. [PMID: 32305250 DOI: 10.1016/j.tibs.2020.03.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022]
Abstract
N-myristoylation (MYR) is a crucial fatty acylation catalyzed by N-myristoyltransferases (NMTs) that is likely to have appeared over 2 billion years ago. Proteome-wide approaches have now delivered an exhaustive list of substrates undergoing MYR across approximately 2% of any proteome, with constituents, several unexpected, associated with different membrane compartments. A set of <10 proteins conserved in eukaryotes probably represents the original set of N-myristoylated targets, marking major changes occurring throughout eukaryogenesis. Recent findings have revealed unexpected mechanisms and reactivity, suggesting competition with other acylations that are likely to influence cellular homeostasis and the steady state of the modification landscape. Here, we review recent advances in NMT catalysis, substrate specificity, and MYR proteomics, and discuss concepts regarding MYR during evolution.
Collapse
Affiliation(s)
- Thierry Meinnel
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| | - Cyril Dian
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Carmela Giglione
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| |
Collapse
|
37
|
Kosciuk T, Price IR, Zhang X, Zhu C, Johnson KN, Zhang S, Halaby SL, Komaniecki GP, Yang M, DeHart CJ, Thomas PM, Kelleher NL, Fromme JC, Lin H. NMT1 and NMT2 are lysine myristoyltransferases regulating the ARF6 GTPase cycle. Nat Commun 2020; 11:1067. [PMID: 32103017 PMCID: PMC7044312 DOI: 10.1038/s41467-020-14893-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/04/2020] [Indexed: 02/08/2023] Open
Abstract
Lysine fatty acylation in mammalian cells was discovered nearly three decades ago, yet the enzymes catalyzing it remain unknown. Unexpectedly, we find that human N-terminal glycine myristoyltransferases (NMT) 1 and 2 can efficiently myristoylate specific lysine residues. They modify ADP-ribosylation factor 6 (ARF6) on lysine 3 allowing it to remain on membranes during the GTPase cycle. We demonstrate that the NAD+-dependent deacylase SIRT2 removes the myristoyl group, and our evidence suggests that NMT prefers the GTP-bound while SIRT2 prefers the GDP-bound ARF6. This allows the lysine myrisotylation-demyristoylation cycle to couple to and promote the GTPase cycle of ARF6. Our study provides an explanation for the puzzling dissimilarity of ARF6 to other ARFs and suggests the existence of other substrates regulated by this previously unknown function of NMT. Furthermore, we identified a NMT/SIRT2-ARF6 regulatory axis, which may offer new ways to treat human diseases.
Collapse
Affiliation(s)
- Tatsiana Kosciuk
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Ian R Price
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Xiaoyu Zhang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Chengliang Zhu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Kayla N Johnson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Shuai Zhang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
- Howard Hughes Medical Institute; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Steve L Halaby
- Department of Molecular Biology and Genetics; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Garrison P Komaniecki
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Min Yang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Caroline J DeHart
- National Resource for Translational and Developmental Proteomics, Departments of Chemistry and Molecular Biosciences and the Feinberg School of Medicine, Northwestern University, Evanston, IL, 60208, USA
| | - Paul M Thomas
- National Resource for Translational and Developmental Proteomics, Departments of Chemistry and Molecular Biosciences and the Feinberg School of Medicine, Northwestern University, Evanston, IL, 60208, USA
| | - Neil L Kelleher
- National Resource for Translational and Developmental Proteomics, Departments of Chemistry and Molecular Biosciences and the Feinberg School of Medicine, Northwestern University, Evanston, IL, 60208, USA
| | - J Christopher Fromme
- Department of Molecular Biology and Genetics; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA.
- Howard Hughes Medical Institute; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
38
|
Kersten C, Fleischer E, Kehrein J, Borek C, Jaenicke E, Sotriffer C, Brenk R. How To Design Selective Ligands for Highly Conserved Binding Sites: A Case Study Using N-Myristoyltransferases as a Model System. J Med Chem 2019; 63:2095-2113. [PMID: 31423787 DOI: 10.1021/acs.jmedchem.9b00586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A model system of two related enzymes with conserved binding sites, namely N-myristoyltransferase from two different organisms, was studied to decipher the driving forces that lead to selective inhibition in such cases. Using a combination of computational and experimental tools, two different selectivity-determining features were identified. For some ligands, a change in side-chain flexibility appears to be responsible for selective inhibition. Remarkably, this was observed for residues orienting their side chains away from the ligands. For other ligands, selectivity is caused by interfering with a water molecule that binds more strongly to the off-target than to the target. On the basis of this finding, a virtual screen for selective compounds was conducted, resulting in three hit compounds with the desired selectivity profile. This study delivers a guideline on how to assess selectivity-determining features in proteins with conserved binding sites and to translate this knowledge into the design of selective inhibitors.
Collapse
Affiliation(s)
- Christian Kersten
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg-Universität Mainz, Staudingerweg 5, 55128 Mainz, Germany.,Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020 Bergen, Norway
| | - Edmond Fleischer
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg-Universität Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | - Josef Kehrein
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020 Bergen, Norway.,Institute of Pharmacy and Food Chemistry, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Christoph Borek
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg-Universität Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | - Elmar Jaenicke
- Institute of Molecular Biophysics, Johannes Gutenberg University, Jakob-Welder-Weg 26, 55128 Mainz, Germany
| | - Christoph Sotriffer
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Ruth Brenk
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020 Bergen, Norway
| |
Collapse
|
39
|
Kallemeijn WW, Lueg GA, Faronato M, Hadavizadeh K, Goya Grocin A, Song OR, Howell M, Calado DP, Tate EW. Validation and Invalidation of Chemical Probes for the Human N-myristoyltransferases. Cell Chem Biol 2019; 26:892-900.e4. [PMID: 31006618 PMCID: PMC6593224 DOI: 10.1016/j.chembiol.2019.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 12/15/2022]
Abstract
On-target, cell-active chemical probes are of fundamental importance in chemical and cell biology, whereas poorly characterized probes often lead to invalid conclusions. Human N-myristoyltransferase (NMT) has attracted increasing interest as target in cancer and infectious diseases. Here we report an in-depth comparison of five compounds widely applied as human NMT inhibitors, using a combination of quantitative whole-proteome N-myristoylation profiling, biochemical enzyme assays, cytotoxicity, in-cell protein synthesis, and cell-cycle assays. We find that N-myristoylation is unaffected by 2-hydroxymyristic acid (100 μM), D-NMAPPD (30 μM), or Tris-DBA palladium (10 μM), with the latter compounds causing cytotoxicity through mechanisms unrelated to NMT. In contrast, drug-like inhibitors IMP-366 (DDD85646) and IMP-1088 delivered complete and specific inhibition of N-myristoylation in a range of cell lines at 1 μM and 100 nM, respectively. This study enables the selection of appropriate on-target probes for future studies and suggests the need for reassessment of previous studies that used off-target compounds.
Collapse
Affiliation(s)
- Wouter W Kallemeijn
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Gregor A Lueg
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Monica Faronato
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kate Hadavizadeh
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Andrea Goya Grocin
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Ok-Ryul Song
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Howell
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Dinis P Calado
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Research Science Hub, 80 Wood Lane, London W12 0BZ, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
40
|
Suwanmanee S, Mahakhunkijcharoen Y, Ampawong S, Leaungwutiwong P, Missé D, Luplertlop N. Inhibition of N-myristoyltransferase1 affects dengue virus replication. Microbiologyopen 2019; 8:e00831. [PMID: 30848105 PMCID: PMC6741125 DOI: 10.1002/mbo3.831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/05/2023] Open
Abstract
Dengue virus (DENV) causes dengue fever, a self‐limiting disease that could be fatal due to serious complications. No specific treatment is currently available and the preventative vaccine is only partially protective. To develop a potential drug target for dengue fever, we need to understand its biology and pathogenesis thoroughly. N‐myristoyltransferase (NMT) is an N‐terminal protein lipidation enzyme that catalyzes the covalent cotranslational attachment of fatty acids to the amino‐terminal glycine residue of a number of proteins, leading to the modulation of various signaling molecules. In this study, we investigated the interaction of dengue viral proteins with host NMT and its subsequent effect on DENV. Our bioinformatics, molecular docking, and far‐western blotting analyses demonstrated the interaction of viral envelope protein (E) with NMT. The gene expression of NMT was strongly elevated in a dependent manner during the viral replication phase in dendritic cells. Moreover, NMT gene silencing significantly inhibited DENV replication in dendritic cells. Further studies investigating the target cell types of other host factors are suggested.
Collapse
Affiliation(s)
- San Suwanmanee
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yuvadee Mahakhunkijcharoen
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Pornsawan Leaungwutiwong
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Dorothée Missé
- MIVEGEC UMR 224, Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Natthanej Luplertlop
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
41
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
42
|
He Y, Liu H, Jiang L, Rui B, Mei J, Xiao H. miR-26 Induces Apoptosis and Inhibits Autophagy in Non-small Cell Lung Cancer Cells by Suppressing TGF-β1-JNK Signaling Pathway. Front Pharmacol 2019; 9:1509. [PMID: 30687089 PMCID: PMC6333751 DOI: 10.3389/fphar.2018.01509] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the causes of cancer mortality worldwide. The role of miR-26 in the development and progression of NSCLC remains largely unknown. In this study we found an abnormal expression of miR-26 in human NSCLC tissues. It was found that miR-26 mimics induced cell apoptosis and promoted caspase-3, 9 activities in human NSCLC cells. The miR-26 inhibitor enhanced the expression of the light chain 3 (LC3) protein and the autophagy related genes in NSCLC cells. Moreover, miR-26 regulated apoptosis and autophagy by inhibiting TGF-β expression in a JNK dependent manner. In addition, miR-26 mimics induced cell apoptosis, was involved in the endoplasmic reticulum stress (ERS) signaling pathway. Down-regulation of the ERS, inhibited apoptosis which was induced by miR-26 mimics in NSCLC cells. In in vivo studies, TUNEL staining revealed that the number of TUNEL positive cells of the tumor tissue in the miR-26 treatment group, were significantly increased in comparison with the control group, while the number of TUNEL positive cells in the tumor tissue were remarkably decreased in the groups treated with miR-26, combined with the TGF-β1 inhibitor or JNK inhibitor. Additionally, the immunoreactivity of TGF-β1 in the cells treated with the miR-26 inhibitor, decreased in comparison to the control group. Our results indicated that miR-26 induced apoptosis and inhibited autophagy in human NSCLC cells through the TGF-β1-JNK signaling pathway, suggesting that miR-26 could be a potential novel target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yi He
- Department of Cardiothoracic Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Liu
- Department of Cardiothoracic Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lianyong Jiang
- Department of Cardiothoracic Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bi Rui
- Department of Cardiothoracic Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ju Mei
- Department of Cardiothoracic Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haibo Xiao
- Department of Cardiothoracic Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
43
|
Goya Grocin A, Serwa RA, Morales Sanfrutos J, Ritzefeld M, Tate EW. Whole Proteome Profiling of N-Myristoyltransferase Activity and Inhibition Using Sortase A. Mol Cell Proteomics 2019; 18:115-126. [PMID: 30341083 PMCID: PMC6317481 DOI: 10.1074/mcp.ra118.001043] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/24/2018] [Indexed: 11/30/2022] Open
Abstract
N-myristoylation is the covalent addition of a 14-carbon saturated fatty acid (myristate) to the N-terminal glycine of specific protein substrates by N-myristoyltransferase (NMT) and plays an important role in protein regulation by controlling localization, stability, and interactions. We developed a novel method for whole-proteome profiling of free N-terminal glycines through labeling with S. Aureus sortase A (SrtA) and used it for assessment of target engagement by an NMT inhibitor. Analysis of the SrtA-labeling pattern with an engineered biotinylated depsipeptide SrtA substrate (Biotin-ALPET-Haa, Haa = 2-hydroxyacetamide) enabled whole proteome identification and quantification of de novo generated N-terminal Gly proteins in response to NMT inhibition by nanoLC-MS/MS proteomics, and was confirmed for specific substrates across multiple cell lines by gel-based analyses and ELISA. To achieve optimal signal over background noise we introduce a novel and generally applicable improvement to the biotin/avidin affinity enrichment step by chemically dimethylating commercial NeutrAvidin resin and combining this with two-step LysC on-bead/trypsin off-bead digestion, effectively eliminating avidin-derived tryptic peptides and enhancing identification of enriched peptides. We also report SrtA substrate specificity in whole-cell lysates for the first time, confirming SrtA promiscuity beyond its recognized preference for N-terminal glycine, and its usefulness as a tool for unbiased labeling of N-terminal glycine-containing proteins. Our new methodology is complementary to metabolic tagging strategies, providing the first approach for whole proteome gain-of signal readout for NMT inhibition in complex samples which are not amenable to metabolic tagging.
Collapse
Affiliation(s)
- Andrea Goya Grocin
- From the ‡Department of Chemistry, Imperial College London, White City Campus, 80 Wood Lane, London W12 0BZ, UK
| | - Remigiusz A Serwa
- From the ‡Department of Chemistry, Imperial College London, White City Campus, 80 Wood Lane, London W12 0BZ, UK
| | - Julia Morales Sanfrutos
- From the ‡Department of Chemistry, Imperial College London, White City Campus, 80 Wood Lane, London W12 0BZ, UK
| | - Markus Ritzefeld
- From the ‡Department of Chemistry, Imperial College London, White City Campus, 80 Wood Lane, London W12 0BZ, UK
| | - Edward W Tate
- From the ‡Department of Chemistry, Imperial College London, White City Campus, 80 Wood Lane, London W12 0BZ, UK..
| |
Collapse
|
44
|
Deng L, Gao X, Liu B, He X, Xu J, Qiang J, Wu Q, Liu S. NMT1 inhibition modulates breast cancer progression through stress-triggered JNK pathway. Cell Death Dis 2018; 9:1143. [PMID: 30446635 PMCID: PMC6240078 DOI: 10.1038/s41419-018-1201-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/11/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Myristoylation is one of key post-translational modifications that involved in signal transduction, cellular transformation and tumorigenesis. Increasing evidence demonstrates that targeting myristoylation might provide a new strategy for eliminating cancers. However, the underlying mechanisms are still yielded unclear. In this study, we demonstrated that genetic inhibition of N-myristoyltransferase NMT1 suppressed initiation, proliferation and invasion of breast cancer cells either in vitro or in vivo. We identified ROS could negatively regulate NMT1 expression and NMT1 knockdown conversely promoted oxidative stress, which formed a feedback loop. Furthermore, inhibition of NMT1 caused degraded proteins increase and ER stress, which cross-talked with mitochondria to produce more ROS. And both of oxidative stress and ER stress could activate JNK pathway, leading to autophagy which abrogated breast cancer progression especially triple-negative breast cancer (TNBC). These studies provide a preclinical proof of concept for targeting NMT1 as a strategy to treat breast cancer.
Collapse
Affiliation(s)
- Lu Deng
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China.,School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xinlei Gao
- School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Bingjie Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China.,School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xueyan He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China
| | - Jiankun Qiang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China
| | - Qingfa Wu
- School of Life Science, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College; Key Laboratory of Breast Cancer in Shanghai; Innovation Center for Cell Signaling Network; Cancer Institutes, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
45
|
Apel C, Bignon J, Garcia-Alvarez MC, Ciccone S, Clerc P, Grondin I, Girard-Valenciennes E, Smadja J, Lopes P, Frédérich M, Roussi F, Meinnel T, Giglione C, Litaudon M. N-myristoyltransferases inhibitory activity of ellagitannins from Terminalia bentzoë (L.) L. f. subsp. bentzoë. Fitoterapia 2018; 131:91-95. [PMID: 30342177 DOI: 10.1016/j.fitote.2018.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/09/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022]
Abstract
N-myristoylation (Myr) is an eukaryotic N-terminal co- or post-translational protein modification in which the enzyme N-myristoyltransferase (NMT) transfers a fatty acid (C14:0) to the N-terminal glycine residues of several cellular key proteins. Depending on the cellular context, NMT may serve as a molecular target in anticancer or anti-infectious therapy, and drugs that inhibit this enzyme may be useful in the treatment of cancer or infectious diseases. As part of an on-going project to identify natural Homo sapiens N-myristoyltransferase 1 inhibitors (HsNMT1), two ellagitannins, punicalagin (1) and isoterchebulin (2), along with eschweilenol C (3) and ellagic acid (4) were isolated from the bark of Terminalia bentzoë (L.) L. f. subsp. bentzoë. Their structures were determined by means of spectroscopic analyses and comparison with literature data. Punicalagin (1) and isoterchebulin (2) showed significant inhibitory activity towards HsNMT1, and also against Plasmodium falciparum NMT (PfNMT) both in vitro and in cellulo, opening alternative paths for new NMT inhibitors development. This is the first report identifying natural products from a botanical source as inhibitors of HsNMT and PfNMT.
Collapse
Affiliation(s)
- Cécile Apel
- Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198 cedex, Gif-sur-Yvette, France.
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198 cedex, Gif-sur-Yvette, France
| | - María Concepción Garcia-Alvarez
- Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198 cedex, Gif-sur-Yvette, France
| | - Sarah Ciccone
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Patricia Clerc
- Laboratoire de Chimie des Substances Naturelles et des Sciences des Aliments (LCSNSA), Université de La Réunion, 15, Avenue rené Cassin, CS 92003-97744 Saint-Denis cedex 9, France
| | - Isabelle Grondin
- Laboratoire de Chimie des Substances Naturelles et des Sciences des Aliments (LCSNSA), Université de La Réunion, 15, Avenue rené Cassin, CS 92003-97744 Saint-Denis cedex 9, France
| | - Emmanuelle Girard-Valenciennes
- Laboratoire de Chimie des Substances Naturelles et des Sciences des Aliments (LCSNSA), Université de La Réunion, 15, Avenue rené Cassin, CS 92003-97744 Saint-Denis cedex 9, France
| | - Jacqueline Smadja
- Laboratoire de Chimie des Substances Naturelles et des Sciences des Aliments (LCSNSA), Université de La Réunion, 15, Avenue rené Cassin, CS 92003-97744 Saint-Denis cedex 9, France
| | - Philippe Lopes
- Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198 cedex, Gif-sur-Yvette, France
| | - Michel Frédérich
- Université de Liège, CIRM (Centre Interfacultaire de Recherche sur le Médicament), Laboratoire de Pharmacognosie, Liège, Belgium
| | - Fanny Roussi
- Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198 cedex, Gif-sur-Yvette, France
| | - Thierry Meinnel
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Carmela Giglione
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France.
| | - Marc Litaudon
- Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198 cedex, Gif-sur-Yvette, France.
| |
Collapse
|
46
|
Health impacts of different edible oils prepared from coconut (Cocos nucifera): A comprehensive review. Trends Food Sci Technol 2018. [DOI: 10.1016/j.tifs.2018.07.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Corbic Ramljak I, Stanger J, Real-Hohn A, Dreier D, Wimmer L, Redlberger-Fritz M, Fischl W, Klingel K, Mihovilovic MD, Blaas D, Kowalski H. Cellular N-myristoyltransferases play a crucial picornavirus genus-specific role in viral assembly, virion maturation, and infectivity. PLoS Pathog 2018; 14:e1007203. [PMID: 30080883 PMCID: PMC6089459 DOI: 10.1371/journal.ppat.1007203] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/13/2018] [Accepted: 07/05/2018] [Indexed: 01/06/2023] Open
Abstract
In nearly all picornaviruses the precursor of the smallest capsid protein VP4 undergoes co-translational N-terminal myristoylation by host cell N-myristoyltransferases (NMTs). Curtailing this modification by mutation of the myristoylation signal in poliovirus has been shown to result in severe assembly defects and very little, if any, progeny virus production. Avoiding possible pleiotropic effects of such mutations, we here used pharmacological abrogation of myristoylation with the NMT inhibitor DDD85646, a pyrazole sulfonamide originally developed against trypanosomal NMT. Infection of HeLa cells with coxsackievirus B3 in the presence of this drug decreased VP0 acylation at least 100-fold, resulting in a defect both early and late in virus morphogenesis, which diminishes the yield of viral progeny by about 90%. Virus particles still produced consisted mainly of provirions containing RNA and uncleaved VP0 and, to a substantially lesser extent, of mature virions with cleaved VP0. This indicates an important role of myristoylation in the viral maturation cleavage. By electron microscopy, these RNA-filled particles were indistinguishable from virus produced under control conditions. Nevertheless, their specific infectivity decreased by about five hundred fold. Since host cell-attachment was not markedly impaired, their defect must lie in the inability to transfer their genomic RNA into the cytosol, likely at the level of endosomal pore formation. Strikingly, neither parechoviruses nor kobuviruses are affected by DDD85646, which appears to correlate with their native capsid containing only unprocessed VP0. Individual knockout of the genes encoding the two human NMT isozymes in haploid HAP1 cells further demonstrated the pivotal role for HsNMT1, with little contribution by HsNMT2, in the virus replication cycle. Our results also indicate that inhibition of NMT can possibly be exploited for controlling the infection by a wide spectrum of picornaviruses.
Collapse
Affiliation(s)
- Irena Corbic Ramljak
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Julia Stanger
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Antonio Real-Hohn
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Dominik Dreier
- Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | - Laurin Wimmer
- Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | | | - Wolfgang Fischl
- Haplogen GmbH, Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | | | - Dieter Blaas
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Heinrich Kowalski
- Center for Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
48
|
Mousnier A, Bell AS, Swieboda DP, Morales-Sanfrutos J, Pérez-Dorado I, Brannigan JA, Newman J, Ritzefeld M, Hutton JA, Guedán A, Asfor AS, Robinson SW, Hopkins-Navratilova I, Wilkinson AJ, Johnston SL, Leatherbarrow RJ, Tuthill TJ, Solari R, Tate EW. Fragment-derived inhibitors of human N-myristoyltransferase block capsid assembly and replication of the common cold virus. Nat Chem 2018; 10:599-606. [PMID: 29760414 PMCID: PMC6015761 DOI: 10.1038/s41557-018-0039-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 03/01/2018] [Indexed: 12/18/2022]
Abstract
Rhinoviruses (RVs) are the pathogens most often responsible for the common cold, and are a frequent cause of exacerbations in asthma, chronic obstructive pulmonary disease and cystic fibrosis. Here we report the discovery of IMP-1088, a picomolar dual inhibitor of the human N-myristoyltransferases NMT1 and NMT2, and use it to demonstrate that pharmacological inhibition of host-cell N-myristoylation rapidly and completely prevents rhinoviral replication without inducing cytotoxicity. The identification of cooperative binding between weak-binding fragments led to rapid inhibitor optimization through fragment reconstruction, structure-guided fragment linking and conformational control over linker geometry. We show that inhibition of the co-translational myristoylation of a specific virus-encoded protein (VP0) by IMP-1088 potently blocks a key step in viral capsid assembly, to deliver a low nanomolar antiviral activity against multiple RV strains, poliovirus and foot and-mouth disease virus, and protection of cells against virus-induced killing, highlighting the potential of host myristoylation as a drug target in picornaviral infections.
Collapse
Affiliation(s)
- Aurélie Mousnier
- National Heart & Lung Institute, Imperial College London, London, UK
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Andrew S Bell
- Department of Chemistry, Imperial College London, London, UK
| | - Dawid P Swieboda
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Inmaculada Pérez-Dorado
- Department of Chemistry, Imperial College London, London, UK
- Department of Life Sciences, Imperial College London, London, UK
| | - James A Brannigan
- Structural Biology Laboratory, Department of Chemistry, University of York, York, UK
| | | | | | - Jennie A Hutton
- Department of Chemistry, Imperial College London, London, UK
| | - Anabel Guedán
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | | | - Iva Hopkins-Navratilova
- Kinetic Discovery Limited, Dundee, UK
- School of Life Sciences, University of Dundee, Dundee, UK
| | - Anthony J Wilkinson
- Structural Biology Laboratory, Department of Chemistry, University of York, York, UK
| | | | - Robin J Leatherbarrow
- Department of Chemistry, Imperial College London, London, UK
- Liverpool John Moores University, Liverpool, UK
| | | | - Roberto Solari
- National Heart & Lung Institute, Imperial College London, London, UK.
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, UK.
| |
Collapse
|
49
|
Schlott AC, Holder AA, Tate EW. N-Myristoylation as a Drug Target in Malaria: Exploring the Role of N-Myristoyltransferase Substrates in the Inhibitor Mode of Action. ACS Infect Dis 2018; 4:449-457. [PMID: 29363940 DOI: 10.1021/acsinfecdis.7b00203] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Malaria continues to be a significant cause of death and morbidity worldwide, and there is a need for new antimalarial drugs with novel targets. We have focused as a potential target for drug development on N-myristoyl transferase (NMT), an enzyme that acylates a wide range of substrate proteins. The NMT substrates in Plasmodium falciparum include some proteins that are common to processes in eukaryotes such as secretory transport and others that are unique to apicomplexan parasites. Myristoylation facilitates a protein interaction with membranes that may be strengthened by further lipidation, and the inhibition of NMT results in incorrect protein localization and the consequent disruption of function. The diverse roles of NMT substrates mean that NMT inhibition has a pleiotropic and severe impact on parasite development, growth, and multiplication. To study the mode of action underlying NMT inhibition, it is important to consider the function of proteins upstream and downstream of NMT. In this work, we therefore present our current perspective on the different functions of known NMT substrates as well as compare the inhibition of cotranslational myristoylation to the inhibition of known targets upstream of NMT.
Collapse
Affiliation(s)
- Anja C. Schlott
- Malaria Parasitology, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom
- Department of Chemistry, Imperial College London, Imperial College Road, SW7 2AZ London, United Kingdom
| | - Anthony A. Holder
- Malaria Parasitology, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom
| | - Edward W. Tate
- Department of Chemistry, Imperial College London, Imperial College Road, SW7 2AZ London, United Kingdom
| |
Collapse
|
50
|
In silico identification of microRNAs predicted to regulate N-myristoyltransferase and Methionine Aminopeptidase 2 functions in cancer and infectious diseases. PLoS One 2018; 13:e0194612. [PMID: 29579063 PMCID: PMC5868815 DOI: 10.1371/journal.pone.0194612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 03/06/2018] [Indexed: 01/16/2023] Open
Abstract
Protein myristoylation is a key protein modification carried out by N-Myristoyltransferase (NMT) after Methionine aminopeptidase 2 (MetAP2) removes methionine from the amino-terminus of the target protein. Protein myristoylation by NMT augments several signaling pathways involved in a myriad of cellular processes, including developmental pathways and pathways that when dysregulated lead to cancer or immune dysfunction. The emerging evidence pointing to NMT-mediated myristoylation as a major cellular regulator underscores the importance of understanding the framework of this type of signaling event. Various studies have investigated the role that myristoylation plays in signaling dysfunction by examining differential gene or protein expression between normal and diseased states, such as cancers or following HIV-1 infection, however no study exists that addresses the role of microRNAs (miRNAs) in the regulation of myristoylation. By performing a large scale bioinformatics and functional analysis of the miRNAs that target key genes involved in myristoylation (NMT1, NMT2, MetAP2), we have narrowed down a list of promising candidates for further analysis. Our condensed panel of miRNAs identifies 35 miRNAs linked to cancer, 21 miRNAs linked to developmental and immune signaling pathways, and 14 miRNAs linked to infectious disease (primarily HIV). The miRNAs panel that was analyzed revealed several NMT-targeting mRNAs (messenger RNA) that are implicated in diseases associated with NMT signaling alteration, providing a link between the realms of miRNA and myristoylation signaling. These findings verify miRNA as an additional facet of myristoylation signaling that must be considered to gain a full perspective. This study provides the groundwork for future studies concerning NMT-transcript-binding miRNAs, and will potentially lead to the development of new diagnostic/prognostic biomarkers and therapeutic targets for several important diseases.
Collapse
|