1
|
Purhonen J, Klefström J, Kallijärvi J. MYC-an emerging player in mitochondrial diseases. Front Cell Dev Biol 2023; 11:1257651. [PMID: 37731815 PMCID: PMC10507175 DOI: 10.3389/fcell.2023.1257651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
The mitochondrion is a major hub of cellular metabolism and involved directly or indirectly in almost all biological processes of the cell. In mitochondrial diseases, compromised respiratory electron transfer and oxidative phosphorylation (OXPHOS) lead to compensatory rewiring of metabolism with resemblance to the Warburg-like metabolic state of cancer cells. The transcription factor MYC (or c-MYC) is a major regulator of metabolic rewiring in cancer, stimulating glycolysis, nucleotide biosynthesis, and glutamine utilization, which are known or predicted to be affected also in mitochondrial diseases. Albeit not widely acknowledged thus far, several cell and mouse models of mitochondrial disease show upregulation of MYC and/or its typical transcriptional signatures. Moreover, gene expression and metabolite-level changes associated with mitochondrial integrated stress response (mt-ISR) show remarkable overlap with those of MYC overexpression. In addition to being a metabolic regulator, MYC promotes cellular proliferation and modifies the cell cycle kinetics and, especially at high expression levels, promotes replication stress and genomic instability, and sensitizes cells to apoptosis. Because cell proliferation requires energy and doubling of the cellular biomass, replicating cells should be particularly sensitive to defective OXPHOS. On the other hand, OXPHOS-defective replicating cells are predicted to be especially vulnerable to high levels of MYC as it facilitates evasion of metabolic checkpoints and accelerates cell cycle progression. Indeed, a few recent studies demonstrate cell cycle defects and nuclear DNA damage in OXPHOS deficiency. Here, we give an overview of key mitochondria-dependent metabolic pathways known to be regulated by MYC, review the current literature on MYC expression in mitochondrial diseases, and speculate how its upregulation may be triggered by OXPHOS deficiency and what implications this has for the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Janne Purhonen
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juha Klefström
- Finnish Cancer Institute, FICAN South Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine, Medical Faculty, University of Helsinki, Helsinki, Finland
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Jukka Kallijärvi
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
2
|
Du Y, Rokavec M, Hermeking H. Squalene epoxidase/SQLE is a candidate target for treatment of colorectal cancers with p53 mutation and elevated c- MYC expression. Int J Biol Sci 2023; 19:4103-4122. [PMID: 37705742 PMCID: PMC10496509 DOI: 10.7150/ijbs.85724] [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: 04/29/2023] [Accepted: 07/30/2023] [Indexed: 09/15/2023] Open
Abstract
Elevated expression of c-MYC and inactivation of p53 represent two of the most common alterations in colorectal cancer (CRC). However, c-MYC and defective p53 are difficult to target therapeutically. Therefore, effectors downstream of both c-MYC and p53 may represent attractive, alternative targets for cancer treatment. In a bioinformatics screen we identified Squalene epoxidase/SQLE as a candidate therapeutic target that appeared to be especially relevant for cell survival in CRCs, which display elevated c-MYC expression and loss of p53 function. SQLE is a rate-limiting enzyme in the cholesterol synthesis. Here, we show that p53 supresses SQLE expression, cholesterol levels, and cell viability via the induction of miR-205, which directly targets SQLE. Furthermore, c-MYC induced SQLE expression directly and via its target gene AP4. The transcription factor AP4/TFAP4 directly induced SQLE expression and cholesterol levels, whereas inactivation of AP4 resulted in decreased SQLE expression and caused resistance to Terbinafine, an inhibitor of SQLE. Inhibition of SQLE decreased viability of CRC cells. This effect was enhanced in CRCs cells with p53 inactivation and/or enhanced c-MYC/AP4 expression. Altogether, our results demonstrate that SQLE represents a vulnerability for CRCs with p53 inactivation and elevated c-MYC activity.
Collapse
Affiliation(s)
- Yuyun Du
- Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-Universität München, D-80337 Munich, Germany
| | - Matjaz Rokavec
- Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-Universität München, D-80337 Munich, Germany
| | - Heiko Hermeking
- Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-Universität München, D-80337 Munich, Germany
- German Cancer Consortium (DKTK), Partner site Munich, D-80336 Munich, Germany
- German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| |
Collapse
|
3
|
Boi D, Rubini E, Breccia S, Guarguaglini G, Paiardini A. When Just One Phosphate Is One Too Many: The Multifaceted Interplay between Myc and Kinases. Int J Mol Sci 2023; 24:4746. [PMID: 36902175 PMCID: PMC10003727 DOI: 10.3390/ijms24054746] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Myc transcription factors are key regulators of many cellular processes, with Myc target genes crucially implicated in the management of cell proliferation and stem pluripotency, energy metabolism, protein synthesis, angiogenesis, DNA damage response, and apoptosis. Given the wide involvement of Myc in cellular dynamics, it is not surprising that its overexpression is frequently associated with cancer. Noteworthy, in cancer cells where high Myc levels are maintained, the overexpression of Myc-associated kinases is often observed and required to foster tumour cells' proliferation. A mutual interplay exists between Myc and kinases: the latter, which are Myc transcriptional targets, phosphorylate Myc, allowing its transcriptional activity, highlighting a clear regulatory loop. At the protein level, Myc activity and turnover is also tightly regulated by kinases, with a finely tuned balance between translation and rapid protein degradation. In this perspective, we focus on the cross-regulation of Myc and its associated protein kinases underlying similar and redundant mechanisms of regulation at different levels, from transcriptional to post-translational events. Furthermore, a review of the indirect effects of known kinase inhibitors on Myc provides an opportunity to identify alternative and combined therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Dalila Boi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisabetta Rubini
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Sara Breccia
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
4
|
Gorachinov F, Mraiche F, Moustafa DA, Hishari O, Ismail Y, Joseph J, Crcarevska MS, Dodov MG, Geskovski N, Goracinova K. Nanotechnology - a robust tool for fighting the challenges of drug resistance in non-small cell lung cancer. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:240-261. [PMID: 36865093 PMCID: PMC9972888 DOI: 10.3762/bjnano.14.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Genomic and proteomic mutation analysis is the standard of care for selecting candidates for therapies with tyrosine kinase inhibitors against the human epidermal growth factor receptor (EGFR TKI therapies) and further monitoring cancer treatment efficacy and cancer development. Acquired resistance due to various genetic aberrations is an unavoidable problem during EGFR TKI therapy, leading to the rapid exhaustion of standard molecularly targeted therapeutic options against mutant variants. Attacking multiple molecular targets within one or several signaling pathways by co-delivery of multiple agents is a viable strategy for overcoming and preventing resistance to EGFR TKIs. However, because of the difference in pharmacokinetics among agents, combined therapies may not effectively reach their targets. The obstacles regarding the simultaneous co-delivery of therapeutic agents at the site of action can be overcome using nanomedicine as a platform and nanotools as delivery agents. Precision oncology research to identify targetable biomarkers and optimize tumor homing agents, hand in hand with designing multifunctional and multistage nanocarriers that respond to the inherent heterogeneity of the tumors, may resolve the challenges of inadequate tumor localization, improve intracellular internalization, and bring advantages over conventional nanocarriers.
Collapse
Affiliation(s)
- Filip Gorachinov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, T6G 2R3 Edmonton, Canada
| | | | - Ola Hishari
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Yomna Ismail
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Jensa Joseph
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Maja Simonoska Crcarevska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Katerina Goracinova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| |
Collapse
|
5
|
Li K, Huo Q, Li BY, Yokota H. The Double-Edged Proteins in Cancer Proteomes and the Generation of Induced Tumor-Suppressing Cells (iTSCs). Proteomes 2023; 11:5. [PMID: 36810561 PMCID: PMC9944087 DOI: 10.3390/proteomes11010005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Unlike a prevalent expectation that tumor cells secrete tumor-promoting proteins and stimulate the progression of neighboring tumor cells, accumulating evidence indicates that the role of tumor-secreted proteins is double-edged and context-dependent. Some of the oncogenic proteins in the cytoplasm and cell membranes, which are considered to promote the proliferation and migration of tumor cells, may inversely act as tumor-suppressing proteins in the extracellular domain. Furthermore, the action of tumor-secreted proteins by aggressive "super-fit" tumor cells can be different from those derived from "less-fit" tumor cells. Tumor cells that are exposed to chemotherapeutic agents could alter their secretory proteomes. Super-fit tumor cells tend to secrete tumor-suppressing proteins, while less-fit or chemotherapeutic agent-treated tumor cells may secrete tumor-promotive proteomes. Interestingly, proteomes derived from nontumor cells such as mesenchymal stem cells and peripheral blood mononuclear cells mostly share common features with tumor cell-derived proteomes in response to certain signals. This review introduces the double-sided functions of tumor-secreted proteins and describes the proposed underlying mechanism, which would possibly be based on cell competition.
Collapse
Affiliation(s)
- Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Qingji Huo
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Weber LI, Hartl M. Strategies to target the cancer driver MYC in tumor cells. Front Oncol 2023; 13:1142111. [PMID: 36969025 PMCID: PMC10032378 DOI: 10.3389/fonc.2023.1142111] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/10/2023] [Indexed: 03/29/2023] Open
Abstract
The MYC oncoprotein functions as a master regulator of cellular transcription and executes non-transcriptional tasks relevant to DNA replication and cell cycle regulation, thereby interacting with multiple proteins. MYC is required for fundamental cellular processes triggering proliferation, growth, differentiation, or apoptosis and also represents a major cancer driver being aberrantly activated in most human tumors. Due to its non-enzymatic biochemical functions and largely unstructured surface, MYC has remained difficult for specific inhibitor compounds to directly address, and consequently, alternative approaches leading to indirect MYC inhibition have evolved. Nowadays, multiple organic compounds, nucleic acids, or peptides specifically interfering with MYC activities are in preclinical or early-stage clinical studies, but none of them have been approved so far for the pharmacological treatment of cancer patients. In addition, specific and efficient delivery technologies to deliver MYC-inhibiting agents into MYC-dependent tumor cells are just beginning to emerge. In this review, an overview of direct and indirect MYC-inhibiting agents and their modes of MYC inhibition is given. Furthermore, we summarize current possibilities to deliver appropriate drugs into cancer cells containing derailed MYC using viral vectors or appropriate nanoparticles. Finding the right formulation to target MYC-dependent cancers and to achieve a high intracellular concentration of compounds blocking or attenuating oncogenic MYC activities could be as important as the development of novel MYC-inhibiting principles.
Collapse
|
7
|
Zhuang JJ, Liu Q, Wu DL, Tie L. Current strategies and progress for targeting the "undruggable" transcription factors. Acta Pharmacol Sin 2022; 43:2474-2481. [PMID: 35132191 PMCID: PMC9525275 DOI: 10.1038/s41401-021-00852-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/23/2021] [Indexed: 11/08/2022] Open
Abstract
Transcription factors (TFs) specifically bind to DNA, recruit cofactor proteins and modulate target gene expression, rendering them essential roles in the regulation of numerous biological processes. Meanwhile, mutated or dysregulated TFs are involved in a variety of human diseases. As multiple signaling pathways ultimately converge at TFs, targeting these TFs directly may prove to be more specific and cause fewer side effects, than targeting the upfront conventional targets in these pathways. All these features together endue TFs with great potential and high selectivity as therapeutic drug targets. However, TFs have been historically considered "undruggable", mainly due to their lack of structural information, especially about the appropriate ligand-binding sites and protein-protein interactions, leading to relatively limited choices in the TF-targeting drug design. In this review, we summarize the recent progress of TF-targeting drugs and highlight certain strategies used for targeting TFs, with a number of representative drugs that have been approved or in the clinical trials as examples. Various approaches in targeting TFs directly or indirectly have been developed. Common direct strategies include aiming at defined binding pockets, proteolysis-targeting chimaera (PROTAC), and mutant protein reactivation. In contrast, the indirect ones comprise inhibition of protein-protein interactions between TF and other proteins, blockade of TF expression, targeting the post-translational modifications, and targeting the TF-DNA interactions. With more comprehensive structural information about TFs revealed by the powerful cryo-electron microscopy technology and predicted by machine-learning algorithms, plus more efficient compound screening platforms and a deeper understanding of TF-disease relationships, the development of TF-targeting drugs will certainly be accelerated in the near future.
Collapse
Affiliation(s)
- Jing-Jing Zhuang
- Marine College, Shandong University, Weihai, 264209, China
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Qian Liu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Da-Lei Wu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
| | - Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China.
| |
Collapse
|
8
|
Nussinov R, Tsai CJ, Jang H. Allostery, and how to define and measure signal transduction. Biophys Chem 2022; 283:106766. [PMID: 35121384 PMCID: PMC8898294 DOI: 10.1016/j.bpc.2022.106766] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/15/2022]
Abstract
Here we ask: What is productive signaling? How to define it, how to measure it, and most of all, what are the parameters that determine it? Further, what determines the strength of signaling from an upstream to a downstream node in a specific cell? These questions have either not been considered or not entirely resolved. The requirements for the signal to propagate downstream to activate (repress) transcription have not been considered either. Yet, the questions are pivotal to clarify, especially in diseases such as cancer where determination of signal propagation can point to cell proliferation and to emerging drug resistance, and to neurodevelopmental disorders, such as RASopathy, autism, attention-deficit/hyperactivity disorder (ADHD), and cerebral palsy. Here we propose a framework for signal transduction from an upstream to a downstream node addressing these questions. Defining cellular processes, experimentally measuring them, and devising powerful computational AI-powered algorithms that exploit the measurements, are essential for quantitative science.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
9
|
Zhu LR, Ni WJ, Cai M, Dai WT, Zhou H. Advances in RNA Epigenetic Modifications in Hepatocellular Carcinoma and Potential Targeted Intervention Strategies. Front Cell Dev Biol 2021; 9:777007. [PMID: 34778277 PMCID: PMC8586511 DOI: 10.3389/fcell.2021.777007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/12/2021] [Indexed: 01/02/2023] Open
Abstract
The current interventions for hepatocellular carcinoma (HCC) are not satisfactory, and more precise targets and promising strategies need to be explored. Recent research has demonstrated the non-negligible roles of RNA epigenetic modifications such as N6-methyladenosine (m6A) and 5-methylcytosine (m5C) in various cancers, including HCC. However, the specific targeting mechanisms are not well elucidated. In this review, we focus on the occurrence and detailed physiopathological roles of multiple RNA modifications on diverse RNAs closely related to the HCC process. In particular, we highlight fresh insights into the impact mechanisms of these posttranscriptional modifications on the whole progression of HCC. Furthermore, we analyzed the possibilities and significance of these modifications and regulators as potential therapeutic targets in HCC treatment, which provides the foundation for exploring targeted intervention strategies. This review will propel the identification of promising therapeutic targets and novel strategies that can be translated into clinical applications for HCC treatment.
Collapse
Affiliation(s)
- Li-Ran Zhu
- Anhui Provincial Children’s Hospital, Anhui Institute of Pediatric Research, Hefei, China
| | - Wei-Jian Ni
- The Key Laboratory of Anti-inflammatory of Immune Medicines, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Ministry of Education, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
- Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Wen-Tao Dai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Key Laboratory of Chinese Medicinal Formula Research, Anhui University of Chinese Medicine, Hefei, China
| | - Hong Zhou
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
10
|
George Kerry R, Ukhurebor KE, Kumari S, Maurya GK, Patra S, Panigrahi B, Majhi S, Rout JR, Rodriguez-Torres MDP, Das G, Shin HS, Patra JK. A comprehensive review on the applications of nano-biosensor-based approaches for non-communicable and communicable disease detection. Biomater Sci 2021; 9:3576-3602. [PMID: 34008586 DOI: 10.1039/d0bm02164d] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The outstretched applications of biosensors in diverse domains has become the reason for their attraction for scientific communities. Because they are analytical devices, they can detect both quantitative and qualitative biological components through the generation of detectable signals. In the recent past, biosensors witnessed significant changes and developments in their design as well as features. Nanotechnology has revolutionized sensing phenomena by increasing biodiagnostic capacity in terms of specificity, size, and cost, resulting in exceptional sensitivity and flexibility. The steep increase of non-communicable diseases across the world has emerged as a matter of concern. In parallel, the abrupt outbreak of communicable diseases poses a serious threat to mankind. For decreasing the morbidity and mortality associated with various communicable and non-communicable diseases, early detection and subsequent treatment are indispensable. Detection of different biological markers generates quantifiable signals that can be electrochemical, mass-based, optical, thermal, or piezoelectric. Speculating on the incumbent applicability and versatility of nano-biosensors in large disciplines, this review highlights different types of biosensors along with their components and detection mechanisms. Moreover, it deals with the current advancements made in biosensors and the applications of nano-biosensors in detection of various non-communicable and communicable diseases, as well as future prospects of nano-biosensors for diagnostics.
Collapse
Affiliation(s)
- Rout George Kerry
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha 751004, India
| | - Kingsley Eghonghon Ukhurebor
- Climatic/Environmental/Telecommunication Unit, Department of Physics, Edo University Iyamho, P.B.M. 04, Auchi, 312101, Edo State, Nigeria
| | - Swati Kumari
- Biopioneer Private limited, Bhubaneswar, Odisha 751024, India
| | - Ganesh Kumar Maurya
- Zoology Section, Mahila MahaVidyalya, Banaras Hindu University, Varanasi-221005, India
| | - Sushmita Patra
- Department of Biotechnology, North Odissa University, Takatpur, Baripada, Odisha 757003, India
| | - Bijayananda Panigrahi
- Biopioneer Private limited, Bhubaneswar, Odisha 751024, India and School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| | - Sanatan Majhi
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha 751004, India
| | | | - María Del Pilar Rodriguez-Torres
- Departamento de Ingeniería Molecular de Materiales, Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Blvd Juriquilla 3001, 76230, Querétaro, Mexico
| | - Gitishree Das
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi, Republic of Korea.
| | - Han-Seung Shin
- Department of Food Science & Biotechnology, Dongguk University-Seoul, Goyangsi, Republic of Korea
| | - Jayanta Kumar Patra
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyangsi, Republic of Korea.
| |
Collapse
|
11
|
Rothzerg E, Xu J, Wood D, Kõks S. 12 Survival-related differentially expressed genes based on the TARGET-osteosarcoma database. Exp Biol Med (Maywood) 2021; 246:2072-2081. [PMID: 33926256 DOI: 10.1177/15353702211007410] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Therapeutically Applicable Research to Generate Effective Treatments (TARGET) project aims to determine molecular changes that drive childhood cancers, including osteosarcoma. The main purpose of the program is to use the open-source database to develop novel, effective, and less toxic therapies. We downloaded TARGET-OS RNA-Sequencing data through R studio and merged the mRNA expression of genes with clinical information (vital status, survival time and gender). Further, we analyzed differential gene expressions between dead and alive patients based on TARGET-OS project. By this study, we found 5758 differentially expressed genes between deceased and alive patients with a false discovery rate below 0.05; 4469 genes were upregulated in deceased patients compared to alive, whereas 1289 genes were downregulated. The survival-related genes were obtained using Kaplan-Meier survival analysis and Cox univariate regression (KM < 0.05 and Cox P-value < 0.05). Out of 5758 differentially expressed genes, only 217 have been associated with overall survival. Eight survival-related downregulated genes (ERCC4, CLUAP1, CTNNBIP1, GCA, RAB40C, SIRPA, USP11, and TCN2) and four survival-related upregulated genes (MUC1, COL13A1, JAG2 and KAZALD1) were selected for further analysis as potential independent prognostic candidate genes. This study may help to discover novel prognostic markers and potential therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia.,Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - David Wood
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
12
|
Fairlie WD, Lee EF. Co-Operativity between MYC and BCL-2 Pro-Survival Proteins in Cancer. Int J Mol Sci 2021; 22:2841. [PMID: 33799592 PMCID: PMC8000576 DOI: 10.3390/ijms22062841] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/30/2022] Open
Abstract
B-Cell Lymphoma 2 (BCL-2), c-MYC and related proteins are arguably amongst the most widely studied in all of biology. Every year there are thousands of papers reporting on different aspects of their biochemistry, cellular and physiological mechanisms and functions. This plethora of literature can be attributed to both proteins playing essential roles in the normal functioning of a cell, and by extension a whole organism, but also due to their central role in disease, most notably, cancer. Many cancers arise due to genetic lesions resulting in deregulation of both proteins, and indeed the development and survival of tumours is often dependent on co-operativity between these protein families. In this review we will discuss the individual roles of both proteins in cancer, describe cancers where co-operativity between them has been well-characterised and finally, some strategies to target these proteins therapeutically.
Collapse
Affiliation(s)
- Walter Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3084, Australia
| | - Erinna F. Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3084, Australia
| |
Collapse
|
13
|
Hartmann O, Reissland M, Maier CR, Fischer T, Prieto-Garcia C, Baluapuri A, Schwarz J, Schmitz W, Garrido-Rodriguez M, Pahor N, Davies CC, Bassermann F, Orian A, Wolf E, Schulze A, Calzado MA, Rosenfeldt MT, Diefenbacher ME. Implementation of CRISPR/Cas9 Genome Editing to Generate Murine Lung Cancer Models That Depict the Mutational Landscape of Human Disease. Front Cell Dev Biol 2021; 9:641618. [PMID: 33738287 PMCID: PMC7961101 DOI: 10.3389/fcell.2021.641618] [Citation(s) in RCA: 21] [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: 12/14/2020] [Accepted: 01/21/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the most common cancer worldwide and the leading cause of cancer-related deaths in both men and women. Despite the development of novel therapeutic interventions, the 5-year survival rate for non-small cell lung cancer (NSCLC) patients remains low, demonstrating the necessity for novel treatments. One strategy to improve translational research is the development of surrogate models reflecting somatic mutations identified in lung cancer patients as these impact treatment responses. With the advent of CRISPR-mediated genome editing, gene deletion as well as site-directed integration of point mutations enabled us to model human malignancies in more detail than ever before. Here, we report that by using CRISPR/Cas9-mediated targeting of Trp53 and KRas, we recapitulated the classic murine NSCLC model Trp53 fl/fl :lsl-KRas G12D/wt . Developing tumors were indistinguishable from Trp53 fl/fl :lsl-KRas G12D/ wt -derived tumors with regard to morphology, marker expression, and transcriptional profiles. We demonstrate the applicability of CRISPR for tumor modeling in vivo and ameliorating the need to use conventional genetically engineered mouse models. Furthermore, tumor onset was not only achieved in constitutive Cas9 expression but also in wild-type animals via infection of lung epithelial cells with two discrete AAVs encoding different parts of the CRISPR machinery. While conventional mouse models require extensive husbandry to integrate new genetic features allowing for gene targeting, basic molecular methods suffice to inflict the desired genetic alterations in vivo. Utilizing the CRISPR toolbox, in vivo cancer research and modeling is rapidly evolving and enables researchers to swiftly develop new, clinically relevant surrogate models for translational research.
Collapse
Affiliation(s)
- Oliver Hartmann
- Deregulated Protein Stability and Cancer Laboratory, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Würzburg, Germany
| | - Michaela Reissland
- Deregulated Protein Stability and Cancer Laboratory, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Würzburg, Germany
| | - Carina R. Maier
- Tumour Metabolism and Microenvironment Group, DKFZ Heidelberg, Heidelberg, Germany
| | - Thomas Fischer
- Deregulated Protein Stability and Cancer Laboratory, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Cristian Prieto-Garcia
- Deregulated Protein Stability and Cancer Laboratory, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Würzburg, Germany
- Faculty of Medicine, TICC, Technion Haifa, Haifa, Israel
| | - Apoorva Baluapuri
- Cancer Systems Biology Group, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
| | - Jessica Schwarz
- Cancer Systems Biology Group, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
| | - Werner Schmitz
- Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
| | - Martin Garrido-Rodriguez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Nikolett Pahor
- Deregulated Protein Stability and Cancer Laboratory, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Würzburg, Germany
| | - Clare C. Davies
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Florian Bassermann
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Amir Orian
- Faculty of Medicine, TICC, Technion Haifa, Haifa, Israel
| | - Elmar Wolf
- Cancer Systems Biology Group, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
| | - Almut Schulze
- Tumour Metabolism and Microenvironment Group, DKFZ Heidelberg, Heidelberg, Germany
| | - Marco A. Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Mathias T. Rosenfeldt
- Mildred Scheel Early Career Center, Würzburg, Germany
- Institut für Pathologie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Markus E. Diefenbacher
- Deregulated Protein Stability and Cancer Laboratory, Lehrstuhl für Biochemie und Molekularbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Würzburg, Germany
| |
Collapse
|
14
|
Shi Q, Xue C, Yuan X, He Y, Yu Z. Gene signatures and prognostic values of m1A-related regulatory genes in hepatocellular carcinoma. Sci Rep 2020; 10:15083. [PMID: 32934298 PMCID: PMC7492257 DOI: 10.1038/s41598-020-72178-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks fourth in cancer-related mortality worldwide. N1-methyladenosine (m1A), a methylation modification on RNA, is gaining attention for its role across diverse biological processes. However, m1A-related regulatory genes expression, its relationship with clinical prognosis, and its role in HCC remain unclear. In this study, we utilized The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) database to investigate alterations within 10 m1A-related regulatory genes and observed a high mutation frequency (23/363). Cox regression analysis and least absolute shrinkage and selection operator were used to explore the association between m1A-related regulatory genes expression and HCC patient survival and identified four regulators that were remarkably associated with HCC patient prognosis. Additionally, an independent cohort from International Cancer Genome Consortium was studied to validate our discoveries and found to be consistent with those in the TCGA dataset. In terms of mechanism, gene set enrichment analysis linked these four genes with various physiological roles in cell division, the MYC pathway, protein metabolism, and mitosis. Kyoto Encyclopedia of Genes and Genomes analysis revealed that PI3K/Akt signaling pathway had potential relevance to m1A-related regulatory genes in HCC. These findings indicate that m1A-related regulatory genes may play crucial roles in regulating HCC progression and be exploited for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Qingmiao Shi
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Chen Xue
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xin Yuan
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yuting He
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China. .,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Zujiang Yu
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China. .,Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
15
|
Sukocheva OA, Furuya H, Ng ML, Friedemann M, Menschikowski M, Tarasov VV, Chubarev VN, Klochkov SG, Neganova ME, Mangoni AA, Aliev G, Bishayee A. Sphingosine kinase and sphingosine-1-phosphate receptor signaling pathway in inflammatory gastrointestinal disease and cancers: A novel therapeutic target. Pharmacol Ther 2020; 207:107464. [PMID: 31863815 DOI: 10.1016/j.pharmthera.2019.107464] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Abstract
Inflammatory gastrointestinal (GI) diseases and malignancies are associated with growing morbidity and cancer-related mortality worldwide. GI tumor and inflammatory cells contain activated sphingolipid-metabolizing enzymes, including sphingosine kinase 1 (SphK1) and SphK2, that generate sphingosine-1-phosphate (S1P), a highly bioactive compound. Many inflammatory responses, including lymphocyte trafficking, are directed by circulatory S1P, present in high concentrations in both the plasma and the lymph of cancer patients. High fat and sugar diet, disbalanced intestinal flora, and obesity have recently been linked to activation of inflammation and SphK/S1P/S1P receptor (S1PR) signaling in various GI pathologies, including cancer. SphK1 overexpression and activation facilitate and enhance the development and progression of esophageal, gastric, and colon cancers. SphK/S1P axis, a mediator of inflammation in the tumor microenvironment, has recently been defined as a target for the treatment of GI disease states, including inflammatory bowel disease and colitis. Several SphK1 inhibitors and S1PR antagonists have been developed as novel anti-inflammatory and anticancer agents. In this review, we analyze the mechanisms of SphK/S1P signaling in GI tissues and critically appraise recent studies on the role of SphK/S1P/S1PR in inflammatory GI disorders and cancers. The potential role of SphK/S1PR inhibitors in the prevention and treatment of inflammation-mediated GI diseases, including GI cancer, is also evaluated.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Hideki Furuya
- Department of Surgery, Samuel Oschin Cancer Center Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mei Li Ng
- Advanced Medical and Dental Institute, University Sains 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Markus Friedemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Vadim V Tarasov
- Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
| | - Vladimir N Chubarev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
| | - Sergey G Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, South Australia 5042, Australia
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia; GALLY International Research Institute, San Antonio, TX 78229, USA; Research Institute of Human Morphology, Moscow 117418, Russia
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
16
|
Hartl M, Puglisi K, Nist A, Raffeiner P, Bister K. The brain acid-soluble protein 1 (BASP1) interferes with the oncogenic capacity of MYC and its binding to calmodulin. Mol Oncol 2020; 14:625-644. [PMID: 31944520 PMCID: PMC7053243 DOI: 10.1002/1878-0261.12636] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 12/16/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
The MYC protein is a transcription factor with oncogenic potential controlling fundamental cellular processes such as cell proliferation, metabolism, differentiation, and apoptosis. The MYC gene is a major cancer driver, and elevated MYC protein levels are a hallmark of most human cancers. We have previously shown that the brain acid-soluble protein 1 gene (BASP1) is specifically downregulated by the v-myc oncogene and that ectopic BASP1 expression inhibits v-myc-induced cell transformation. The 11-amino acid effector domain of the BASP1 protein interacts with the calcium sensor calmodulin (CaM) and is mainly responsible for this inhibitory function. We also reported recently that CaM interacts with all MYC variant proteins and that ectopic CaM increases the transactivation and transformation potential of the v-Myc protein. Here, we show that the presence of excess BASP1 or of a synthetic BASP1 effector domain peptide leads to displacement of v-Myc from CaM. The protein stability of v-Myc is decreased in cells co-expressing v-Myc and BASP1, which may account for the inhibition of v-Myc. Furthermore, suppression of v-Myc-triggered transcriptional activation and cell transformation is compensated by ectopic CaM, suggesting that BASP1-mediated withdrawal of CaM from v-Myc is a crucial event in the inhibition. In view of the tumor-suppressive role of BASP1 which was recently also reported for human cancer, small compounds or peptides based on the BASP1 effector domain could be used in drug development strategies aimed at tumors with high MYC expression.
Collapse
Affiliation(s)
- Markus Hartl
- Institute of Biochemistry and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Kane Puglisi
- Institute of Biochemistry and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Andrea Nist
- Institute of Biochemistry and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Philipp Raffeiner
- Institute of Biochemistry and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Klaus Bister
- Institute of Biochemistry and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| |
Collapse
|
17
|
Sukocheva OA, Lukina E, McGowan E, Bishayee A. Sphingolipids as mediators of inflammation and novel therapeutic target in inflammatory bowel disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 120:123-158. [PMID: 32085881 DOI: 10.1016/bs.apcsb.2019.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Morbidity of inflammatory gastrointestinal (GI) diseases continues to grow resulting in worsen quality of life and increased burden on public medical systems. Complex and heterogenous illnesses, inflammatory bowel diseases (IBDs) encompass several inflammation -associated pathologies including Crohn's disease and ulcerative colitis. IBD is often initiated by a complex interplay between host genetic and environmental factors, lifestyle and diet, and intestinal bacterial components. IBD inflammatory signature was linked to the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) signaling pathway that is currently targeted by IBD therapies. Sphingolipid signaling was identified as one of the key mediators and regulators of pro-inflammatory conditions, and, specifically, TNF-α related signaling. All GI tissues and circulating immune/blood cells contain activated sphingolipid-metabolizing enzymes, including sphingosine kinases (SphK1 and SphK2) that generate sphingosine-1-phosphate (S1P), a bioactive lipid and ligand for five G-protein coupled membrane S1P receptors (S1PRs). Numerous normal and pathogenic inflammatory responses are mediated by SphK/S1P/S1PRs signaling axis including lymphocyte trafficking and activation of cytokine signaling machinery. SphK1/S1P/S1PRs axis has recently been defined as a target for the treatment of GI diseases including IBD/colitis. Several SphK1 inhibitors and S1PRs antagonists have been developed as novel anti-inflammatory agents. In this review, we discuss the mechanisms of SphK/S1P signaling in inflammation-linked GI disorders. The potential role of SphK/S1PRs inhibitors in the prevention and treatment of IBD/colitis is critically evaluated.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Elena Lukina
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Eileen McGowan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
18
|
Gutiérrez-González M, Latorre Y, Zúñiga R, Aguillón JC, Molina MC, Altamirano C. Transcription factor engineering in CHO cells for recombinant protein production. Crit Rev Biotechnol 2019; 39:665-679. [PMID: 31030575 DOI: 10.1080/07388551.2019.1605496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The continuous increase of approved biopharmaceutical products drives the development of more efficient recombinant protein expression systems. Chinese hamster ovary (CHO) cells are the mainstay for this purpose but have some drawbacks, such as low levels of expression. Several strategies have been applied to increase the productivity of CHO cells with different outcomes. Transcription factor (TF) engineering has emerged as an interesting and successful approach, as these proteins can act as master regulators; the expression and function of a TF can be controlled by small molecules, and it is possible to design tailored TFs and promoters with desired features. To date, the majority of studies have focused on the use of TFs with growth, metabolic, cell cycle or endoplasmic reticulum functions, although there is a trend to develop new, synthetic TFs. Moreover, new synthetic biological approaches are showing promising advances for the development of specific TFs, even with tailored ligand sensitivity. In this article, we summarize the strategies to increase recombinant protein expression by modulating and designing TFs and with advancements in synthetic biology. We also illustrate how this class of proteins can be used to develop more robust expression systems.
Collapse
Affiliation(s)
| | - Yesenia Latorre
- b Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Roberto Zúñiga
- a Centro de InmunoBiotecnología, Universidad de Chile , Santiago , Chile
| | | | | | - Claudia Altamirano
- b Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| |
Collapse
|
19
|
Abstract
The intestinal epithelium is a multicellular interface in close proximity to a dense microbial milieu that is completely renewed every 3-5 days. Pluripotent stem cells reside at the crypt, giving rise to transient amplifying cells that go through continuous steps of proliferation, differentiation and finally anoikis (a form of programmed cell death) while migrating upwards to the villus tip. During these cellular transitions, intestinal epithelial cells (IECs) possess distinct metabolic identities reflected by changes in mitochondrial activity. Mitochondrial function emerges as a key player in cell fate decisions and in coordinating cellular metabolism, immunity, stress responses and apoptosis. Mediators of mitochondrial signalling include molecules such as ATP and reactive oxygen species and interrelate with pathways such as the mitochondrial unfolded protein response (MT-UPR) and AMP kinase signalling, in turn affecting cell cycle progression and stemness. Alterations in mitochondrial function and MT-UPR activation are integral aspects of pathologies, including IBD and cancer. Mitochondrial signalling and concomitant changes in metabolism contribute to intestinal homeostasis and regulate IEC dedifferentiation-differentiation programmes in the context of diseases, suggesting that mitochondrial function as a cellular checkpoint critically contributes to disease outcome. This Review highlights mitochondrial function and MT-UPR signalling in epithelial cell stemness, differentiation and lineage commitment and illustrates mitochondrial function in intestinal diseases.
Collapse
|
20
|
Burkholderia Lethal Factor 1, a Novel Anti-Cancer Toxin, Demonstrates Selective Cytotoxicity in MYCN-Amplified Neuroblastoma Cells. Toxins (Basel) 2018; 10:toxins10070261. [PMID: 29954071 PMCID: PMC6071135 DOI: 10.3390/toxins10070261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022] Open
Abstract
Immunotoxins are being investigated as anti-cancer therapies and consist of a cytotoxic enzyme fused to a cancer targeting antibody. All currently used toxins function via the inhibition of protein synthesis, making them highly potent in both healthy and transformed cells. This non-specific cell killing mechanism causes dose-limiting side effects that can severely limit the potential of immunotoxin therapy. In this study, the recently characterised bacterial toxin Burkholderia lethal factor 1 (BLF1) is investigated as a possible alternative payload for targeted toxin therapy in the treatment of neuroblastoma. BLF1 inhibits translation initiation by inactivation of eukaryotic initiation translation factor 4A (eIF4A), a putative anti-cancer target that has been shown to regulate a number of oncogenic proteins at the translational level. We show that cellular delivery of BLF1 selectively induces apoptosis in neuroblastoma cells that display MYCN amplification but has little effect on non-transformed cells. Future immunotoxins based on this enzyme may therefore have higher specificity towards MYCN-amplified cancer cells than more conventional ribosome-inactivating proteins, leading to an increased therapeutic window and decreased side effects.
Collapse
|
21
|
MYC overexpression with its prognostic and clinicopathological significance in breast cancer. Oncotarget 2017; 8:93998-94008. [PMID: 29212204 PMCID: PMC5706850 DOI: 10.18632/oncotarget.21501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022] Open
Abstract
Background Proto-oncogene MYC has been indicated to promote progression of many cancers. However, prognostic and clinicopathological significance of MYC in breast cancer need further evaluation. Methods We searched EMBASE and PubMed databases to find useful studies. We analyzed relationships between high MYC expression and prognostic data/ clinicopathological features through hazard ratio (HR) and odds ratio (OR). Each statistical test was two-sided. Results There were 29 studies (36 cohorts) with 12621 patients enrolled in our study The MYC overexpression was associated with worse DFS/RFS (disease/relapse free survival) in 11 studies (16 cohorts) with 5390 patients, and OS (overall survival) of 7 studies (8 cohorts) with 2672 patients. Subgroup analysis according to ethnicity/technique/data source displayed that MYC overexpression was associated with poor DFS/RFS in FISH, other technique, all data source and Asian/Non-Asian subgroup, and worse OS in all subgroups. In addition, MYC overexpression was related to large tumor size, high histologic grade, lymph node metastasis, negative hormone receptors and positive Ki67 expression. Conclusions Our results showed that MYC overexpression was associated with worse prognosis and high risk of breast cancer, especially in patients with negative hormone receptors, which highlighted the potential of MYC as a significant prognostic biomarker of breast cancer.
Collapse
|
22
|
Nickkholgh B, Sittadjody S, Rothberg MB, Fang X, Li K, Chou JW, Hawkins GA, Balaji K. Beta-catenin represses protein kinase D1 gene expression by non-canonical pathway through MYC/MAX transcription complex in prostate cancer. Oncotarget 2017; 8:78811-78824. [PMID: 29108267 PMCID: PMC5668000 DOI: 10.18632/oncotarget.20229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/09/2017] [Indexed: 12/13/2022] Open
Abstract
Down regulation of Protein Kinase D1 (PrKD1), a novel serine threonine kinase, in prostate, gastric, breast and colon cancers in humans leads to disease progression. While the down regulation of PrKD1 by DNA methylation in gastric cancer and by nuclear beta-catenin in colon cancer has been shown, the regulatory mechanisms in other cancers are unknown. Because we had demonstrated that PrKD1 is the only known kinase to phosphorylate threonine 120 (T120) of beta-catenin in prostate cancer resulting in increased nuclear beta-catenin, we explored the role of beta-catenin in gene regulation of PrKD1. An initial CHIP assay identified potential binding sites for beta-catenin in and downstream of PrKD1 promoter and sequencing confirmed recruitment of beta-catenin to a 166 base pairs sequence upstream of exon 2. Co-transfection studies with PrKD1-promoter-reporter suggested that beta-catenin represses PrKD1 promoter. Efforts to identify transcription factors that mediate the co-repressor effects of beta-catenin identified recruitment of both MYC and its obligate heterodimer MAX to the same binding site as beta-catenin on the PrKD1 promoter site. Moreover, treatment with MYC inhibitor rescued the co-repressor effect of beta-catenin on PrKD1 gene expression. Prostate specific knock out of PrKD1 in transgenic mice demonstrated increased nuclear expression of beta-catenin validating the in vitro studies. Functional studies showed that nuclear translocation of beta-catenin as a consequence of PrKD1 down regulation, increases AR transcriptional activity with attendant downstream effects on androgen responsive genes. In silico human gene expression analysis confirmed the down regulation of PrKD1 in metastatic prostate cancer correlated inversely with the expression of MAX, but not MYC, and positively with MXD1, a competing heterodimer of MAX, suggesting that the dimerization of MAX with either MYC or MXD1 regulates PrKD1 gene expression. The study has identified a novel auto-repressive loop that perpetuates PrKD1 down regulation through beta-catenin/MYC/MAX protein complex.
Collapse
Affiliation(s)
- Bita Nickkholgh
- Department of Physiology-Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Wake Forest Institute for Regenerative Medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sivanandane Sittadjody
- Wake Forest Institute for Regenerative Medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Xiaolan Fang
- Wake Forest Institute for Regenerative Medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Cancer Biology, School of Medicine, Wake Forest University, Winston-Salem, NC, USA
- Current/Present address: Clinical Bioinformatics, New York Genome Center, New York, NY, USA
| | - Kunzhao Li
- Biology Department, Wake Forest University, Winston-Salem, NC, USA
| | - Jeff W. Chou
- Department of Biostatistical Sciences, Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Gregory A. Hawkins
- Center for Genomics and Personalized Medicine and WFB Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - K.C. Balaji
- Wake Forest Institute for Regenerative Medicine (WFRM), Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Urology, Wake Forest Baptist Health, Winston Salem, NC, USA
- W.G.(Bill) Hefner Veterans Administration Medical Center, Salisbury, NC, USA
- Department of Cancer Biology, School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
23
|
Fernandez-Pol S, De Stefano D, Kim J. Immunohistochemistry reveals an increased proportion of MYC-positive cells in subcutaneous panniculitis-like T-cell lymphoma compared with lupus panniculitis. J Cutan Pathol 2017; 44:925-930. [PMID: 28800143 DOI: 10.1111/cup.13025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/17/2017] [Accepted: 08/07/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND Subcutaneous panniculitis-like T-cell lymphoma (SPTCL) is a malignant primary cutaneous T-cell lymphoma that shares significant clinical, histopathologic and immunophenotypic overlap with lupus erythematosus panniculitis (LEP). METHODS We performed immunohistochemistry for the MYC oncoprotein on 23 cases of SPTCL (1 CD8 negative) and 12 cases of LEP to evaluate if there are quantitative or qualitative differences in protein expression of this marker in these entities. RESULTS In SPTCL cases, the percentage of all cells that were c-Myc positive ranged from 0.8% to 16%, with a mean of 5.0% and a median of 4.4%. In contrast, in the LEP cases, the percentage of c-Myc-positive cells in the cases ranged from 0.34% to 3.7%, averaged 1.4% and the median was 0.8%. The difference between the means of these 2 diagnostic categories was statistically significant. Fluorescence in situ hybridization performed on 4 cases of SPTCL with a relatively high level of MYC immunohistochemical staining, however, failed to demonstrate evidence of MYC rearrangement or amplification. CONCLUSIONS Our work demonstrates that MYC expression levels differ between these 2 histologic mimics and suggests that this important oncoprotein may play a role in the pathogenesis of SPTCL.
Collapse
Affiliation(s)
| | - Danielle De Stefano
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Jinah Kim
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
24
|
Fallah Y, Brundage J, Allegakoen P, Shajahan-Haq AN. MYC-Driven Pathways in Breast Cancer Subtypes. Biomolecules 2017; 7:biom7030053. [PMID: 28696357 PMCID: PMC5618234 DOI: 10.3390/biom7030053] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/19/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022] Open
Abstract
The transcription factor MYC (MYC proto-oncogene, bHLH transcription factor) is an essential signaling hub in multiple cellular processes that sustain growth of many types of cancers. MYC regulates expression of RNA, both protein and non-coding, that control central metabolic pathways, cell death, proliferation, differentiation, stress pathways, and mechanisms of drug resistance. Activation of MYC has been widely reported in breast cancer progression. Breast cancer is a complex heterogeneous disease and treatment options are primarily guided by histological and biochemical evaluations of the tumors. Based on biochemical markers, three main breast cancer categories are ER+ (estrogen receptor alpha positive), HER2+ (human epidermal growth factor receptor 2 positive), and TNBC (triple-negative breast cancer; estrogen receptor negative, progesterone receptor negative, HER2 negative). MYC is elevated in TNBC compared with other cancer subtypes. Interestingly, MYC-driven pathways are further elevated in aggressive breast cancer cells and tumors that display drug resistant phenotype. Identification of MYC target genes is essential in isolating signaling pathways that drive tumor development. In this review, we address the role of MYC in the three major breast cancer subtypes and highlight the most promising leads to target MYC functions.
Collapse
Affiliation(s)
- Yassi Fallah
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Janetta Brundage
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Paul Allegakoen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
25
|
Song W, Hwang Y, Youngblood VM, Cook RS, Balko JM, Chen J, Brantley-Sieders DM. Targeting EphA2 impairs cell cycle progression and growth of basal-like/triple-negative breast cancers. Oncogene 2017; 36:5620-5630. [PMID: 28581527 PMCID: PMC5629103 DOI: 10.1038/onc.2017.170] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 02/06/2023]
Abstract
Basal-like/triple-negative breast cancers (TNBCs) are among the most aggressive forms of breast cancer, and disproportionally affects young premenopausal women and women of African descent. Patients with TNBC suffer a poor prognosis due in part to a lack of molecularly targeted therapies, which represents a critical barrier for effective treatment. Here, we identify EphA2 receptor tyrosine kinase as a clinically relevant target for TNBC. EphA2 expression is enriched in the basal-like molecular subtype in human breast cancers. Loss of EphA2 function in both human and genetically engineered mouse models of TNBC reduced tumor growth in culture and in vivo. Mechanistically, targeting EphA2 impaired cell cycle progression through S-phase via downregulation of c-Myc and stabilization of the cyclin-dependent kinase inhibitor p27/KIP1. A small molecule kinase inhibitor of EphA2 effectively suppressed tumor cell growth in vivo, including TNBC patient-derived xenografts. Thus, our data identify EphA2 as a novel molecular target for TNBC.
Collapse
Affiliation(s)
- W Song
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Y Hwang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - V M Youngblood
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - R S Cook
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J M Balko
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J Chen
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Cellular and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - D M Brantley-Sieders
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
26
|
Zaytseva O, Quinn LM. Controlling the Master: Chromatin Dynamics at the MYC Promoter Integrate Developmental Signaling. Genes (Basel) 2017; 8:genes8040118. [PMID: 28398229 PMCID: PMC5406865 DOI: 10.3390/genes8040118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/15/2017] [Accepted: 04/07/2017] [Indexed: 02/06/2023] Open
Abstract
The transcription factor and cell growth regulator MYC is potently oncogenic and estimated to contribute to most cancers. Decades of attempts to therapeutically target MYC directly have not resulted in feasible clinical applications, and efforts have moved toward indirectly targeting MYC expression, function and/or activity to treat MYC-driven cancer. A multitude of developmental and growth signaling pathways converge on the MYC promoter to modulate transcription through their downstream effectors. Critically, even small increases in MYC abundance (<2 fold) are sufficient to drive overproliferation; however, the details of how oncogenic/growth signaling networks regulate MYC at the level of transcription remain nebulous even during normal development. It is therefore essential to first decipher mechanisms of growth signal-stimulated MYC transcription using in vivo models, with intact signaling environments, to determine exactly how these networks are dysregulated in human cancer. This in turn will provide new modalities and approaches to treat MYC-driven malignancy. Drosophila genetic studies have shed much light on how complex networks signal to transcription factors and enhancers to orchestrate Drosophila MYC (dMYC) transcription, and thus growth and patterning of complex multicellular tissue and organs. This review will discuss the many pathways implicated in patterning MYC transcription during development and the molecular events at the MYC promoter that link signaling to expression. Attention will also be drawn to parallels between mammalian and fly regulation of MYC at the level of transcription.
Collapse
Affiliation(s)
- Olga Zaytseva
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
- School of Biomedical Sciences, University of Melbourne, Parkville 3010, Australia.
| | - Leonie M Quinn
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
- School of Biomedical Sciences, University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
27
|
Whitfield JR, Beaulieu ME, Soucek L. Strategies to Inhibit Myc and Their Clinical Applicability. Front Cell Dev Biol 2017; 5:10. [PMID: 28280720 PMCID: PMC5322154 DOI: 10.3389/fcell.2017.00010] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/03/2017] [Indexed: 12/20/2022] Open
Abstract
Myc is an oncogene deregulated in most-perhaps all-human cancers. Each Myc family member, c-, L-, and N-Myc, has been connected to tumor progression and maintenance. Myc is recognized as a "most wanted" target for cancer therapy, but has for many years been considered undruggable, mainly due to its nuclear localization, lack of a defined ligand binding site, and physiological function essential to the maintenance of normal tissues. The challenge of identifying a pharmacophore capable of overcoming these hurdles is reflected in the current absence of a clinically-viable Myc inhibitor. The first attempts to inhibit Myc used antisense technology some three decades ago, followed by small molecule inhibitors discovered through "classical" compound library screens. Notable breakthroughs proving the feasibility of systemic Myc inhibition were made with the Myc dominant negative mutant Omomyc, showing both the great promise in targeting this infamous oncogene for cancer treatment as well as allaying fears about the deleterious side effects that Myc inhibition might have on normal proliferating tissues. During this time many other strategies have appeared in an attempt to drug the undruggable, including direct and indirect targeting, knockdown, protein/protein and DNA interaction inhibitors, and translation and expression regulation. The inhibitors range from traditional small molecules to natural chemicals, to RNA and antisense, to peptides and miniproteins. Here, we briefly describe the many approaches taken so far, with a particular focus on their potential clinical applicability.
Collapse
Affiliation(s)
- Jonathan R Whitfield
- Vall d'Hebron Institute of Oncology, Edifici Cellex, Hospital Vall d'Hebron Barcelona, Spain
| | | | - Laura Soucek
- Vall d'Hebron Institute of Oncology, Edifici Cellex, Hospital Vall d'HebronBarcelona, Spain; Peptomyc, Edifici Cellex, Hospital Vall d'HebronBarcelona, Spain; Institució Catalana de Recerca i Estudis AvançatsBarcelona, Spain; Department of Biochemistry and Molecular Biology, Universitat Autònoma de BarcelonaBellaterra, Spain
| |
Collapse
|
28
|
Dozzo M, Carobolante F, Donisi PM, Scattolin A, Maino E, Sancetta R, Viero P, Bassan R. Burkitt lymphoma in adolescents and young adults: management challenges. Adolesc Health Med Ther 2017; 8:11-29. [PMID: 28096698 PMCID: PMC5207020 DOI: 10.2147/ahmt.s94170] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
About one-half of all Burkitt lymphoma (BL) patients are younger than 40 years, and one-third belong to the adolescent and young adult (AYA) subset, defined by an age between 15 and 25-40 years, based on selection criteria used in different reports. BL is an aggressive B-cell neoplasm displaying highly characteristic clinico-diagnostic features, the biologic hallmark of which is a translocation involving immunoglobulin and c-MYC genes. It presents as sporadic, endemic, or epidemic disease. Endemicity is pathogenetically linked to an imbalance of the immune system which occurs in African children infected by malaria parasites and Epstein-Barr virus, while the epidemic form strictly follows the pattern of infection by HIV. BL shows propensity to extranodal involvement of abdominal organs, bone marrow, and central nervous system, and can cause severe metabolic and renal impairment. Nevertheless, BL is highly responsive to specifically designed short-intensive, rotational multiagent chemotherapy programs, empowered by the anti-CD20 monoclonal antibody rituximab. When carefully applied with appropriate supportive measures, these modern programs achieve a cure rate of approximately 90% in the average AYA patient, irrespective of clinical stage, which is the best result achievable in any aggressive lymphoid malignancy to date. The challenges ahead concern the following: optimization of management in underdeveloped countries, with reduction of diagnostic and referral-for-care intervals, and the applicability of currently curative regimens; the development of lower intensity but equally effective treatments for frail or immunocompromised patients at risk of death by complications; the identification of very high-risk patients through positron-emission tomography and minimal residual disease assays; and the assessment in these and the few refractory/relapsed ones of new monoclonals (ofatumumab, blinatumomab, inotuzumab ozogamicin) and new molecules targeting c-MYC and key proliferative steps of B-cell malignancies.
Collapse
Affiliation(s)
- Massimo Dozzo
- Complex Operative Unit of Hematology, Ospedale dell’Angelo
| | | | - Pietro Maria Donisi
- Simple Departmental Operative Unit of Anatomic Pathology, Ospedale Ss. Giovanni e Paolo, Venice, Italy
| | | | - Elena Maino
- Complex Operative Unit of Hematology, Ospedale dell’Angelo
| | | | - Piera Viero
- Complex Operative Unit of Hematology, Ospedale dell’Angelo
| | - Renato Bassan
- Complex Operative Unit of Hematology, Ospedale dell’Angelo
- Correspondence: Renato Bassan, Complex Operative Unit of Hematology, Ospedale dell’Angelo, Via Paccagnella 11, 30174 Mestre-Venice, Italy, Tel +39 41 965 7362, Fax +39 41 965 7361, Email
| |
Collapse
|