1
|
Cartwright M, Jha RK, Smith AT. Structure and Mechanism of Aminoacyl-tRNA-Protein L/F- and R-transferases. J Mol Biol 2025; 437:169210. [PMID: 40381981 DOI: 10.1016/j.jmb.2025.169210] [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: 04/03/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The aminoacyl-tRNA-protein transferases (also known as aa-transferases) are a class of enzymes that utilize a highly conserved GCN5-related N-acetyltransferase (GNAT) fold to catalyze the post-translational transfer of amino acids from an aminoacylated transfer RNA (tRNA) to an acceptor protein. The two most important subclasses of aa-transferases are the prokaryotic L/F-transferases and the eukaryotic R-transferases (ATE1s). Both subclasses were initially discovered as early as the 1960s, and both share an overlapping function linked to protein degradation: L/F-transferases are known to modify proteins that are ultimately targeted for degradation via the Clp proteolytic pathway, while R-transferases (ATE1s) are known to modify proteins that may be targeted for degradation by the ubiquitin proteasome system (UPS), although many non-degradative fates may also occur. While L/F-transferases have been minimally explored at the cellular level, the R-transferases (ATE1s) have had extensive studies linking them to critical cellular functions. Despite over a half a century passing since their discoveries, X-ray crystallographic and cryo-EM studies have only recently begun to shed light onto the mechanism of these enzymes. This review underscores the functional importance of L/F- and R-transferases (ATE1s) and highlights the recent structural developments in this field with a particular emphasis on the eukaryotic R-transferases (ATE1s). Additionally, this review draws on current structural information to synopsize proposed catalytic and regulatory mechanisms for these enzymes. Finally, this review highlights important structural and mechanistic knowledge gaps in aa-transferase function that should be addressed in order to target these important enzymes for future therapeutic developments.
Collapse
Affiliation(s)
- Misti Cartwright
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Rajat Kumar Jha
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Aaron T Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
2
|
Sogbein O, Paul P, Umar M, Chaari A, Batuman V, Upadhyay R. Bortezomib in cancer therapy: Mechanisms, side effects, and future proteasome inhibitors. Life Sci 2024; 358:123125. [PMID: 39413903 DOI: 10.1016/j.lfs.2024.123125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
The ubiquitin-proteasome pathway (UPP) regulates protein stability and normal cellular functions with the help of autocatalytic proteasome complex. Studies have linked aberrant proteasome activity to malignant cells and found that proteasome inhibitors play a significant role as therapeutic drugs for various types of cancer, specifically multiple myeloma and mantle cell lymphoma. Bortezomib, the first FDA-approved proteasome inhibitor for treating different stages of multiple myeloma, acts on cancer cells by inhibiting the 26S proteasome, modulating NF-κB, phosphorylating Bcl-2, upregulating of NOXA, blocking p53 degradation, activating caspase, generating reactive oxygen species (ROS), and inhibiting angiogenesis. However, its efficacy is limited due to side effects such as peripheral neuropathy (PN), thrombotic microangiopathy (TMA), and acute interstitial nephritis (AIN). Therefore, a better understanding of its precise mechanism of action may help mitigate these side effects. In this review, we have discussed the proposed mechanisms of action and off target effects of Bortezomib, along with the prospects of next generation potential proteasome inhibitor drugs in the treatment of cancer.
Collapse
Affiliation(s)
- Olusola Sogbein
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Meenakshi Umar
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Vecihi Batuman
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Rohit Upadhyay
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
3
|
Baran O, Akgun MY, Kayhan A, Evran S, Ozbek A, Akyoldas G, Samanci MY, Demirel N, Sonmez D, Serin H, Kocak A, Kemerdere R, Tanriverdi T. The association between calreticulin and glucagon-like peptide-1 expressions with prognostic factors in high-grade gliomas. J Cancer Res Ther 2024; 20:25-32. [PMID: 38554294 DOI: 10.4103/jcrt.jcrt_1519_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/26/2022] [Indexed: 04/01/2024]
Abstract
OBJECTIVE The aim of this study is to present the expressions of Calreticulin (CALR) and Glucagon-like peptide-1 (GLP-1) in high-grade gliomas and to further show the relation between the levels of these molecules and Ki-67 index, presence of Isocitrate dehydrogenase (IDH)-1 mutation, and tumor grade. PATIENTS AND METHODS A total of 43 patients who underwent surgical resection due to high-grade gliomas (HGG) (grades III and IV) were included. The control group comprised 27 people who showed no gross pathology in the brain during the autopsy procedures. Adequately sized tumor samples were removed from each patient during surgery, and cerebral tissues were removed from the control subjects during the autopsy procedures. Each sample was stored at -80°C as rapidly as possible until the enzyme assay. RESULTS Patients with high-grade gliomas showed significantly higher levels of CALR and significantly lower levels of GLP-1 when compared to control subjects (P = 0.001). CALR levels were significantly higher, GLP-1 levels were significantly lower in grade IV gliomas than those in grade III gliomas (P = 0.001). Gliomas with negative IDH-1 mutations had significantly higher CALR expressions and gliomas with positive IDH-1 mutations showed significantly higher GLP-1 expressions (P = 0.01). A positive correlation between Ki-67 and CALR and a negative correlation between Ki-67 and GLP-1 expressions were observed in grade IV gliomas (P = 0.001). CONCLUSIONS Our results showed that higher CALR and lower GLP-1 expressions are found in HGGs compared to normal cerebral tissues.
Collapse
Affiliation(s)
- Oguz Baran
- Department of Neurosurgery, Koç University Hospital, Istanbul, Turkey
| | | | - Ahmet Kayhan
- Department of Neurosurgery, Haseki Research and Training Hospital, Istanbul, Turkey
| | - Sevket Evran
- Department of Neurosurgery, Haseki Research and Training Hospital, Istanbul, Turkey
| | - Arif Ozbek
- Department of Neurosurgery, Medipol Mega University Hospital, Istanbul, Turkey
| | - Goktug Akyoldas
- Department of Neurosurgery, Koç University Hospital, Istanbul, Turkey
| | | | - Nail Demirel
- Department of Neurosurgery, Istanbul Research and Training Hospital, Istanbul, Turkey
| | - Derya Sonmez
- Clinical Biochemistry Laboratory, Istanbul Research and Training Hospital, Istanbul, Turkey
| | - Huriye Serin
- Clinical Biochemistry Laboratory, Istanbul Research and Training Hospital, Istanbul, Turkey
| | - Ayhan Kocak
- Department of Neurosurgery, Taksim Research and Training Hospital, Istanbul, Turkey
| | - Rahsan Kemerdere
- Department of Neurosurgery, Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Taner Tanriverdi
- Department of Neurosurgery, Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
4
|
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
|
5
|
Abeywansha T, Huang W, Ye X, Nawrocki A, Lan X, Jankowsky E, Taylor DJ, Zhang Y. The structural basis of tRNA recognition by arginyl-tRNA-protein transferase. Nat Commun 2023; 14:2232. [PMID: 37076488 PMCID: PMC10115844 DOI: 10.1038/s41467-023-38004-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 04/03/2023] [Indexed: 04/21/2023] Open
Abstract
Arginyl-tRNA-protein transferase 1 (ATE1) is a master regulator of protein homeostasis, stress response, cytoskeleton maintenance, and cell migration. The diverse functions of ATE1 arise from its unique enzymatic activity to covalently attach an arginine onto its protein substrates in a tRNA-dependent manner. However, how ATE1 (and other aminoacyl-tRNA transferases) hijacks tRNA from the highly efficient ribosomal protein synthesis pathways and catalyzes the arginylation reaction remains a mystery. Here, we describe the three-dimensional structures of Saccharomyces cerevisiae ATE1 with and without its tRNA cofactor. Importantly, the putative substrate binding domain of ATE1 adopts a previously uncharacterized fold that contains an atypical zinc-binding site critical for ATE1 stability and function. The unique recognition of tRNAArg by ATE1 is coordinated through interactions with the major groove of the acceptor arm of tRNA. Binding of tRNA induces conformational changes in ATE1 that helps explain the mechanism of substrate arginylation.
Collapse
Affiliation(s)
- Thilini Abeywansha
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Xuan Ye
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Allison Nawrocki
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Xin Lan
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Eckhard Jankowsky
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Derek J Taylor
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Yi Zhang
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
6
|
Li Y, Jiang JX, Fan W, Fish SR, Das S, Gupta P, Mozes G, Vancza L, Sarkar S, Kunimoto K, Chen D, Park H, Clemens D, Tomilov A, Cortopassi G, Török NJ. Shc Is Implicated in Calreticulin-Mediated Sterile Inflammation in Alcoholic Hepatitis. Cell Mol Gastroenterol Hepatol 2022; 15:197-211. [PMID: 36122677 PMCID: PMC9676381 DOI: 10.1016/j.jcmgh.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Src homology and collagen (Shc) proteins are major adapters to extracellular signals, however, the regulatory role of Shc isoforms in sterile inflammatory responses in alcoholic hepatitis (AH) has not been fully investigated. We hypothesized that in an isoform-specific manner Shc modulates pre-apoptotic signals, calreticulin (CRT) membrane exposure, and recruitment of inflammatory cells. METHODS Liver biopsy samples from patients with AH vs healthy subjects were studied for Shc expression using DNA microarray data and immunohistochemistry. Shc knockdown (hypomorph) and age-matched wild-type mice were pair-fed according to the chronic-plus-binge alcohol diet. To analyze hepatocyte-specific effects, adeno-associated virus 8-thyroxine binding globulin-Cre (hepatocyte-specific Shc knockout)-mediated deletion was performed in flox/flox Shc mice. Lipid peroxidation, proinflammatory signals, redox radicals, reduced nicotinamide adenine dinucleotide/oxidized nicotinamide adenine dinucleotide ratio, as well as cleaved caspase 8, B-cell-receptor-associated protein 31 (BAP31), Bcl-2-associated X protein (Bax), and Bcl-2 homologous antagonist killer (Bak), were assessed in vivo. CRT translocation was studied in ethanol-exposed p46ShcẟSH2-transfected hepatocytes by membrane biotinylation in conjunction with phosphorylated-eukaryotic initiation factor 2 alpha, BAP31, caspase 8, and Bax/Bak. The effects of idebenone, a novel Shc inhibitor, was studied in alcohol/pair-fed mice. RESULTS Shc was significantly induced in patients with AH (P < .01). Alanine aminotransferase, reduced nicotinamide adenine dinucleotide/oxidized nicotinamide adenine dinucleotide ratios, production of redox radicals, and lipid peroxidation improved (P < .05), and interleukin 1β, monocyte chemoattractant protein 1, and C-X-C chemokine ligand 10 were reduced in Shc knockdown and hepatocyte-specific Shc knockout mice. In vivo, Shc-dependent induction, and, in hepatocytes, a p46Shc-dependent increase in pre-apoptotic proteins Bax/Bak, caspase 8, BAP31 cleavage, and membrane translocation of CRT/endoplasmic reticulum-resident protein 57 were seen. Idebenone protected against alcohol-mediated liver injury. CONCLUSIONS Alcohol induces p46Shc-dependent activation of pre-apoptotic pathways and translocation of CRT to the membrane, where it acts as a damage-associated molecular pattern, instigating immunogenicity. Shc inhibition could be a novel treatment strategy in AH.
Collapse
Affiliation(s)
- Yuan Li
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Joy X Jiang
- Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, California
| | - Weiguo Fan
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Sarah R Fish
- Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, California
| | - Suvarthi Das
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Parul Gupta
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Gergely Mozes
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Lorand Vancza
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Sutapa Sarkar
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Koshi Kunimoto
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Dongning Chen
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Hyesuk Park
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California
| | - Dahn Clemens
- Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Alexey Tomilov
- Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California
| | - Gino Cortopassi
- Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California
| | - Natalie J Török
- Gastroenterology and Hepatology, Stanford University, VA Palo Alto, Palo Alto, California.
| |
Collapse
|
7
|
Asadi MR, Moslehian MS, Sabaie H, Poornabi M, Ghasemi E, Hassani M, Hussen BM, Taheri M, Rezazadeh M. Stress Granules in the Anti-Cancer Medications Mechanism of Action: A Systematic Scoping Review. Front Oncol 2021; 11:797549. [PMID: 35004322 PMCID: PMC8739770 DOI: 10.3389/fonc.2021.797549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/08/2021] [Indexed: 12/16/2022] Open
Abstract
Stress granule (SG) formation is a well-known cellular mechanism for minimizing stress-related damage and increasing cell survival. In addition to playing a critical role in the stress response, SGs have emerged as critical mediators in human health. It seems logical that SGs play a key role in cancer cell formation, development, and metastasis. Recent studies have shown that many SG components contribute to the anti-cancer medications' responses through tumor-associated signaling pathways and other mechanisms. SG proteins are known for their involvement in the translation process, control of mRNA stability, and capacity to function in both the cytoplasm and nucleus. The current systematic review aimed to include all research on the impact of SGs on the mechanism of action of anti-cancer medications and was conducted using a six-stage methodological framework and the PRISMA guideline. Prior to October 2021, a systematic search of seven databases for eligible articles was performed. Following the review of the publications, the collected data were subjected to quantitative and qualitative analysis. Notably, Bortezomib, Sorafenib, Oxaliplatin, 5-fluorouracil, Cisplatin, and Doxorubicin accounted for the majority of the medications examined in the studies. Overall, this systematic scoping review attempts to demonstrate and give a complete overview of the function of SGs in the mechanism of action of anti-cancer medications by evaluating all research.
Collapse
Affiliation(s)
- Mohammad Reza Asadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hani Sabaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziye Poornabi
- Student Research Committee, School of Medicine, Shahroud University of Medical Science, Shahroud, Iran
| | - Elham Ghasemi
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mehdi Hassani
- Student Research Committee, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Maryam Rezazadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Chen L, Kashina A. Post-translational Modifications of the Protein Termini. Front Cell Dev Biol 2021; 9:719590. [PMID: 34395449 PMCID: PMC8358657 DOI: 10.3389/fcell.2021.719590] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Post-translational modifications (PTM) involve enzyme-mediated covalent addition of functional groups to proteins during or after synthesis. These modifications greatly increase biological complexity and are responsible for orders of magnitude change between the variety of proteins encoded in the genome and the variety of their biological functions. Many of these modifications occur at the protein termini, which contain reactive amino- and carboxy-groups of the polypeptide chain and often are pre-primed through the actions of cellular machinery to expose highly reactive residues. Such modifications have been known for decades, but only a few of them have been functionally characterized. The vast majority of eukaryotic proteins are N- and C-terminally modified by acetylation, arginylation, tyrosination, lipidation, and many others. Post-translational modifications of the protein termini have been linked to different normal and disease-related processes and constitute a rapidly emerging area of biological regulation. Here we highlight recent progress in our understanding of post-translational modifications of the protein termini and outline the role that these modifications play in vivo.
Collapse
Affiliation(s)
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
9
|
Seo T, Kim J, Shin HC, Kim JG, Ju S, Nawale L, Han G, Lee HS, Bang G, Kim JY, Bang JK, Lee KH, Soung NK, Hwang J, Lee C, Kim SJ, Kim BY, Cha-Molstad H. R-catcher, a potent molecular tool to unveil the arginylome. Cell Mol Life Sci 2021; 78:3725-3741. [PMID: 33687501 PMCID: PMC8038991 DOI: 10.1007/s00018-021-03805-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/18/2021] [Accepted: 02/27/2021] [Indexed: 11/27/2022]
Abstract
Protein arginylation is a critical regulator of a variety of biological processes. The ability to uncover the global arginylation pattern and its associated signaling pathways would enable us to identify novel disease targets. Here, we report the development of a tool able to capture the N-terminal arginylome. This tool, termed R-catcher, is based on the ZZ domain of p62, which was previously shown to bind N-terminally arginylated proteins. Mutating the ZZ domain enhanced its binding specificity and affinity for Nt-Arg. R-catcher pulldown coupled to LC-MS/MS led to the identification of 59 known and putative arginylated proteins. Among these were a subgroup of novel ATE1-dependent arginylated ER proteins that are linked to diverse biological pathways, including cellular senescence and vesicle-mediated transport as well as diseases, such as Amyotrophic Lateral Sclerosis and Alzheimer's disease. This study presents the first molecular tool that allows the unbiased identification of arginylated proteins, thereby unlocking the arginylome and provide a new path to disease biomarker discovery.
Collapse
Affiliation(s)
- Taewook Seo
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jihyo Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea
| | - Ho-Chul Shin
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jung Gi Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Shinyeong Ju
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Laxman Nawale
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Goeun Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea.,Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hye Seon Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28116, Republic of Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28116, Republic of Korea
| | - Jeong Kyu Bang
- Division of Magnetic Resonance, Korea Basic Science Institute, Ochang, 28116, Republic of Korea
| | - Kyung Ho Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea
| | - Joonsung Hwang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea
| | - Cheolju Lee
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seung Jun Kim
- Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea. .,Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
| | - Bo Yeon Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea. .,Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Hyunjoo Cha-Molstad
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang-eup, Cheongju-si, Chungcheongbuk-do, 28116, Republic of Korea. .,Department of Biomolecular Science, KRIBB School, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
10
|
Su Z, Han S, Jin Q, Zhou N, Lu J, Shangguan F, Yu S, Liu Y, Wang L, Lu J, Li Q, Cai L, Wang C, Tian X, Chen L, Zheng W, Lu B. Ciclopirox and bortezomib synergistically inhibits glioblastoma multiforme growth via simultaneously enhancing JNK/p38 MAPK and NF-κB signaling. Cell Death Dis 2021; 12:251. [PMID: 33674562 PMCID: PMC7935936 DOI: 10.1038/s41419-021-03535-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 02/05/2023]
Abstract
Ciclopirox (CPX) is an antifungal drug that has recently been reported to act as a potential anticancer drug. However, the effects and underlying molecular mechanisms of CPX on glioblastoma multiforme (GBM) remain unknown. Bortezomib (BTZ) is the first proteasome inhibitor-based anticancer drug approved to treat multiple myeloma and mantle cell lymphoma, as BTZ exhibits toxic effects on diverse tumor cells. Herein, we show that CPX displays strong anti-tumorigenic activity on GBM. Mechanistically, CPX inhibits GBM cellular migration and invasion by reducing N-Cadherin, MMP9 and Snail expression. Further analysis revealed that CPX suppresses the expression of several key subunits of mitochondrial enzyme complex, thus leading to the disruption of mitochondrial oxidative phosphorylation (OXPHOS) in GBM cells. In combination with BTZ, CPX promotes apoptosis in GBM cells through the induction of reactive oxygen species (ROS)-mediated c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK) signaling. Moreover, CPX and BTZ synergistically activates nuclear factor kappa B (NF-κB) signaling and induces cellular senescence. Our findings suggest that a combination of CPX and BTZ may serve as a novel therapeutic strategy to enhance the anticancer activity of CPX against GBM.
Collapse
Affiliation(s)
- Zhipeng Su
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shengnan Han
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Pathology, The Second Hospital of Jiaxing, Jiaxing University, Jiaxing, 314000, China
| | - Qiumei Jin
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ningning Zhou
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Junwan Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fugen Shangguan
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shiyi Yu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yongzhang Liu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lu Wang
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jianglong Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Li
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lin Cai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chengde Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaohe Tian
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and molecular imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Lingyan Chen
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Weiming Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Bin Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
11
|
Wagner PM, Prucca CG, Velazquez FN, Sosa Alderete LG, Caputto BL, Guido ME. Temporal regulation of tumor growth in nocturnal mammals: In vivo studies and chemotherapeutical potential. FASEB J 2021; 35:e21231. [PMID: 33428275 DOI: 10.1096/fj.202001753r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/28/2020] [Accepted: 11/12/2020] [Indexed: 11/11/2022]
Abstract
Tumors of the nervous system including glioblastoma multiforme (GBM) are the most frequent and aggressive form of brain tumors; however, little is known about the impact of the circadian timing system on the formation, growth, and treatment of these tumors. We investigated day/night differences in tumor growth after injection of A530 glioma cells isolated from malignant peripheral nerve sheath tumor (MPNSTs) of NPcis (Trp53+/- ; Nf1+/- ) mice. Synchronized A530 cell cultures expressing typical glial markers were injected at the beginning of the day or night into the sciatic nerve zone of C57BL/6 mice subject to a 12:12 hours light/dark (LD) cycle or after being released to constant darkness (DD). Tumors generated in animals injected early at night in the LD cycle or in DD showed higher growth rates than in animals injected diurnally. No differences were found when animals were injected at the same time with cultures synchronized 12 hours apart. Similar experiments performed with B16 melanoma cells showed higher tumor growth rates in animals injected at the beginning of the night compared to those injected in the daytime. A higher tumor growth rate than that in controls was observed when mice were injected with knocked-down clock gene Bmal1 cells. Finally, when we compared day/night administration of different doses of the proteasome inhibitor Bortezomib (0.5-1.5 mg/kg) in tumor-bearing animals, we found that low-dose chemotherapy displayed higher efficacy when administered at night. Results suggest the existence of a precise temporal control of tumor growth and of drug efficacy in which the host state and susceptibility are critical.
Collapse
Affiliation(s)
- Paula M Wagner
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - César G Prucca
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fabiola N Velazquez
- Stony Brook Cancer Center and the Department of Medicine,, Stony Brook University, Stony Brook, USA
| | - Lucas G Sosa Alderete
- Instituto de Biotecnología Ambiental y Salud (INBIAS, UNRC-CONICET). Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, Argentina
| | - Beatriz L Caputto
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mario E Guido
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
12
|
Van V, Smith AT. ATE1-Mediated Post-Translational Arginylation Is an Essential Regulator of Eukaryotic Cellular Homeostasis. ACS Chem Biol 2020; 15:3073-3085. [PMID: 33228359 DOI: 10.1021/acschembio.0c00677] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Arginylation is a protein post-translational modification catalyzed by arginyl-tRNA transferases (ATE1s), which are critical enzymes conserved across all eukaryotes. Arginylation is a key step in the Arg N-degron pathway, a hierarchical cellular signaling pathway that links the ubiquitin-dependent degradation of a protein to the identity of its N-terminal amino acid side chain. The fidelity of ATE1-catalyzed arginylation is imperative, as this post-translational modification regulates several essential biological processes such as cardiovascular maturation, chromosomal segregation, and even the stress response. While the process of ATE1-catalyzed arginylation has been studied in detail at the cellular level, much remains unknown about the structure of this important enzyme, its mechanism of action, and its regulation. In this work, we detail the current state of knowledge on ATE1-catalyzed arginylation, and we discuss both ongoing and future directions that will reveal the structural and mechanistic details of this essential eukaryotic cellular regulator.
Collapse
Affiliation(s)
- Verna Van
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Aaron T. Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
13
|
Avcilar-Kucukgoze I, Kashina A. Hijacking tRNAs From Translation: Regulatory Functions of tRNAs in Mammalian Cell Physiology. Front Mol Biosci 2020; 7:610617. [PMID: 33392265 PMCID: PMC7773854 DOI: 10.3389/fmolb.2020.610617] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Transfer tRNAs (tRNAs) are small non-coding RNAs that are highly conserved in all kingdoms of life. Originally discovered as the molecules that deliver amino acids to the growing polypeptide chain during protein synthesis, tRNAs have been believed for a long time to play exclusive role in translation. However, recent studies have identified key roles for tRNAs and tRNA-derived small RNAs in multiple other processes, including regulation of transcription and translation, posttranslational modifications, stress response, and disease. These emerging roles suggest that tRNAs may be central players in the complex machinery of biological regulatory pathways. Here we overview these non-canonical roles of tRNA in normal physiology and disease, focusing largely on eukaryotic and mammalian systems.
Collapse
Affiliation(s)
- Irem Avcilar-Kucukgoze
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
14
|
Wagner PM, Monjes NM, Guido ME. Chemotherapeutic Effect of SR9009, a REV-ERB Agonist, on the Human Glioblastoma T98G Cells. ASN Neuro 2020; 11:1759091419892713. [PMID: 31825658 PMCID: PMC6909277 DOI: 10.1177/1759091419892713] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma multiforme is the most aggressive brain tumor, and human T98G cells constitute a useful glioblastoma multiforme model to evaluate the chemotherapeutic agents. Modern life (shiftwork, jetlag, etc.) may cause circadian disorganization promoting higher cancer risk and metabolic disorders. Although little is known about the tumor-intrinsic circadian clock function, pharmacological modulation of circadian components may offer selective anticancer strategies. REV-ERBs are heme-binding circadian clock components acting as repressors of processes involved in tumorigenesis such as metabolism, proliferation, and inflammation. A synthetic pyrrole derivative (SR9009) that acts as REV-ERBs-specific agonists exhibits potent in vivo activity on metabolism and tumor cell viability. Here, we investigated SR9009 effects on T98G cell viability, differential chemotherapy time responses, and underlying metabolic processes (reactive oxygen species [ROS] and lipid droplets [LDs]) and compared it with the proteasome inhibitor Bortezomib treatment. SR9009-treated cells exhibited significant reduction in cell viability with consequences on cell cycle progression. Dexamethasone synchronized cells displayed differential time responses to SR9009 treatment with highest responses 18 to 30 h after synchronization. SR9009 treatment decreased ROS levels while Bortezomib increased them. However, both treatments significantly increased LD levels, whereas the combined treatment showed additive or synergistic effects between both drugs. In addition, we extended these studies to HepG2 cells which also showed a significant decrease in cell viability and ROS levels and the increase in LD levels after SR9009 treatment. Our results suggest that the pharmacological modulation of the tumor-intrinsic clock by REV-ERB agonists severely affects cell metabolism and promotes cytotoxic effects on cancer cells.
Collapse
Affiliation(s)
- Paula M Wagner
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto," Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia M Monjes
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto," Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mario E Guido
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto," Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
15
|
Abstract
Calreticulin (CRT) is a pleiotropic and highly conserved molecule that is mainly localized in the endoplasmic reticulum. Recently, CRT has gained special interest for its functions outside the endoplasmic reticulum where it has immunomodulatory properties. CRT translocation to the cell membrane serves as an "eat me" signal and promotes efferocytosis of apoptotic cells and cancer cell removal with completely opposite outcomes. Efferocytosis results in a silenced immune response and homeostasis, while removal of dying cancer cells brought about by anthracycline treatment, ionizing-irradiation or photodynamic therapy results in immunogenic cell death with activation of the innate and adaptive immune responses. In addition, CRT impacts phagocyte activation and cytokine production. The effects of CRT on cytokine production depend on its conformation, species specificity, degree of oligomerization and/or glycosylation, as well as its cellular localization and the molecular partners involved. The controversial roles of CRT in cancer progression and the possible role of the CALR gene mutations in myeloproliferative neoplasms are also addressed. The release of CRT and its influence on the different cells involved during efferocytosis and immunogenic cell death points to additional roles of CRT besides merely acting as an "eat me" signal during apoptosis. Understanding the contribution of CRT in physiological and pathological processes could give us some insight into the potential of CRT as a therapeutic target.
Collapse
|
16
|
Wang J, Ren D, Sun Y, Xu C, Wang C, Cheng R, Wang L, Jia G, Ren J, Ma J, Tu Y, Ji H. Inhibition of PLK4 might enhance the anti-tumour effect of bortezomib on glioblastoma via PTEN/PI3K/AKT/mTOR signalling pathway. J Cell Mol Med 2020; 24:3931-3947. [PMID: 32126150 PMCID: PMC7171416 DOI: 10.1111/jcmm.14996] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/26/2019] [Accepted: 11/23/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma (GBM) is one of the most common aggressive cancers of the central nervous system in adults with a high mortality rate. Bortezomib is a boronic acid-based potent proteasome inhibitor that has been actively studied for its anti-tumour effects through inhibition of the proteasome. The proteasome is a key component of the ubiquitin-proteasome pathway that is critical for protein homeostasis, regulation of cellular growth, and apoptosis. Overexpression of polo-like kinase 4 (PLK4) is commonly reported in tumour cells and increases their invasive and metastatic abilities. In this study, we established a cell model of PLK4 knockdown and overexpression in LN-18, A172 and LN-229 cells and found that knockdown of PLK4 expression enhanced the anti-tumour effect of bortezomib. We further found that this effect may be mediated by the PTEN/PI3K/AKT/mTOR signalling pathway and that the apoptotic and oxidative stress processes were activated, while the expression of matrix metalloproteinases (MMPs) was down-regulated. Similar phenomenon was observed using in vitro experiments. Thus, we speculate that PLK4 inhibition may be a new therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Jing Wang
- Department of neurosurgery, Shanxi academy of medical science, Shanxi Bethune Hospital, Taiyuan, China
| | - Dengpeng Ren
- Department of neurosurgery, Central Hospital of Yuncheng city, Yuncheng, China
| | - Yan Sun
- Neurological intensive care unit, Special medical center of PAP, Tianjin, China
| | - Chao Xu
- Neurological intensive care unit, Special medical center of PAP, Tianjin, China
| | - Chunhong Wang
- Department of neurosurgery, Shanxi people's hospital, Taiyuan, China
| | - Rui Cheng
- Department of neurosurgery, Shanxi people's hospital, Taiyuan, China
| | - Lina Wang
- Neurological intensive care unit, Special medical center of PAP, Tianjin, China
| | - Guijun Jia
- Department of neurosurgery, Shanxi people's hospital, Taiyuan, China
| | - Jinrui Ren
- Department of neurosurgery, Shanxi people's hospital, Taiyuan, China
| | - Jiuhong Ma
- Department of neurosurgery, Shanxi people's hospital, Taiyuan, China
| | - Yue Tu
- Neurological intensive care unit, Special medical center of PAP, Tianjin, China
| | - Hongming Ji
- Department of neurosurgery, Shanxi people's hospital, Taiyuan, China
| |
Collapse
|