1
|
He Y, Zhou J, Gao H, Liu C, Zhan P, Liu X. Broad-spectrum antiviral strategy: Host-targeting antivirals against emerging and re-emerging viruses. Eur J Med Chem 2024; 265:116069. [PMID: 38160620 DOI: 10.1016/j.ejmech.2023.116069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Viral infections are amongst the most prevalent diseases that pose a significant threat to human health. Targeting viral proteins or host factors represents two primary strategies for the development of antiviral drugs. In contrast to virus-targeting antivirals (VTAs), host-targeting antivirals (HTAs) offer advantages in terms of overcoming drug resistance and effectively combating a wide range of viruses, including newly emerging ones. Therefore, targeting host factors emerges as an extremely promising strategy with the potential to address critical challenges faced by VTAs. In recent years, extensive research has been conducted on the discovery and development of HTAs, leading to the approval of maraviroc, a chemokine receptor type 5 (CCR5) antagonist used for the treatment of HIV-1 infected individuals, with several other potential treatments in various stages of development for different viral infections. This review systematically summarizes advancements made in medicinal chemistry regarding various host targets and classifies them into four distinct catagories based on their involvement in the viral life cycle: virus attachment and entry, biosynthesis, nuclear import and export, and viral release.
Collapse
Affiliation(s)
- Yong He
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Jiahui Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Huizhan Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, Shandong Province, PR China.
| |
Collapse
|
2
|
Gotur D, Case A, Liu J, Sickmier EA, Holt N, Knockenhauer KE, Yao S, Lee YT, Copeland RA, Buker SM, Boriack-Sjodin PA. Development of assays to support identification and characterization of modulators of DExH-box helicase DHX9. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:376-384. [PMID: 37625785 DOI: 10.1016/j.slasd.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
DHX9 is a DExH-box RNA helicase that utilizes hydrolysis of all four nucleotide triphosphates (NTPs) to power cycles of 3' to 5' directional movement to resolve and/or unwind double stranded RNA, DNA, and RNA/DNA hybrids, R-loops, triplex-DNA and G-quadraplexes. DHX9 activity is important for both viral amplification and maintaining genomic stability in cancer cells; therefore, it is a therapeutic target of interest for drug discovery efforts. Biochemical assays measuring ATP hydrolysis and oligonucleotide unwinding for DHX9 have been developed and characterized, and these assays can support high-throughput compound screening efforts under balanced conditions. Assay development efforts revealed DHX9 can use double stranded RNA with 18-mer poly(U) 3' overhangs and as well as significantly shorter overhangs at the 5' or 3' end as substrates. The enzymatic assays are augmented by a robust SPR assay for compound validation. A mechanism-derived inhibitor, GTPγS, was characterized as part of the validation of these assays and a crystal structure of GDP bound to cat DHX9 has been solved. In addition to enabling drug discovery efforts for DHX9, these assays may be extrapolated to other RNA helicases providing a valuable toolkit for this important target class.
Collapse
Affiliation(s)
- Deepali Gotur
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - April Case
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - Julie Liu
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - E Allen Sickmier
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - Nicholas Holt
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | | | - Shihua Yao
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | - Young-Tae Lee
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | | | - Shane M Buker
- Accent Therapeutics, 1050 Waltham Street, Lexington, MA 02421, USA
| | | |
Collapse
|
3
|
Lin TC. Perturbation Analysis of a Prognostic DDX3X-Mediated Gene Expression Signature Identifies the Antimetastatic Potential of Chaetocin in Hepatocellular Carcinoma. Cells 2023; 12:1628. [PMID: 37371098 DOI: 10.3390/cells12121628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
ATP-dependent RNA helicase DDX3X, also known as DEAD (Asp-Glu-Ala-Asp) Box Polypeptide 3, X-Linked (DDX3X), is critical for RNA metabolism, and emerging evidence implicates ATP-dependent RNA helicase DDX3X's participation in various cellular processes to modulate cancer progression. In this study, the clinical significance of DDX3X was addressed, and DDX3X was identified as a biomarker for poor prognosis. An exploration of transcriptomic data from 373 liver cancer patients from The Cancer Genome Atlas (TCGA) using Ingenuity Pathway Analysis (IPA) suggested an association between DDX3X expression and cancer metastasis. Lentiviral-based silencing of DDX3X in a hepatocellular carcinoma (HCC) cell line resulted in the suppression of cell migration and invasion. The molecular mechanism regarding ATP-dependent RNA helicase DDX3X in liver cancer progression had been addressed in many studies. I focused on the biological application of the DDX3X-mediated gene expression signature in cancer therapeutics. An investigation of the DDX3X-correlated expression signature via the L1000 platform of Connectivity Map (BROAD Institute) first identified a histone methyltransferase inhibitor, chaetocin, as a novel compound for alleviating metastasis in HCC. In this study, the prognostic value of DDX3X and the antimetastatic property of chaetocin are presented to shed light on the development of anti-liver cancer strategies.
Collapse
Affiliation(s)
- Tsung-Chieh Lin
- Genomic Medicine Core Laboratory, Department of Medical Research and Development, Chang Gung Memorial Hospital, Linkou, Taoyuan City 333, Taiwan
- Department of Biomedical Sciences, Chang Gung University, Taoyuan City 333, Taiwan
| |
Collapse
|
4
|
Heaton SM, Gorry PR, Borg NA. DExD/H-box helicases in HIV-1 replication and their inhibition. Trends Microbiol 2023; 31:393-404. [PMID: 36463019 DOI: 10.1016/j.tim.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022]
Abstract
Antiretroviral therapy (ART) reduces human immunodeficiency virus type 1 (HIV-1) infection, but selection of treatment-refractory variants remains a major challenge. HIV-1 encodes 16 canonical proteins, a small number of which are the singular targets of nearly all antiretrovirals developed to date. Cellular factors are increasingly being explored, which may present more therapeutic targets, more effectively target certain aspects of the viral replication cycle, and/or limit viral escape. Unlike most other positive-sense RNA viruses that encode at least one helicase, retroviruses are limited to the host repertoire. Accordingly, HIV-1 subverts DEAD-box helicase 3X (DDX3X) and numerous other cellular helicases of the Asp-Glu-x-Asp/His (DExD/H)-box family to service multiple aspects of its replication cycle. Here we review DDX3X and other DExD/H-box helicases in HIV-1 replication and their inhibition.
Collapse
Affiliation(s)
- Steven M Heaton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia; Current affiliation: RIKEN Cluster for Pioneering Research and RIKEN Center for Integrative Medical Sciences, 1-chōme-7-22 Suehirochō, Tsurumi-ku, Yokohama 230-0045, Kanagawa, Japan.
| | - Paul R Gorry
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Natalie A Borg
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| |
Collapse
|
5
|
Naineni SK, Robert F, Nagar B, Pelletier J. Targeting DEAD-box RNA helicases: The emergence of molecular staples. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1738. [PMID: 35581936 DOI: 10.1002/wrna.1738] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/29/2022]
Abstract
RNA helicases constitute a large family of proteins that play critical roles in mediating RNA function. They have been implicated in all facets of gene expression pathways involving RNA, from transcription to processing, transport and translation, and storage and decay. There is significant interest in developing small molecule inhibitors to RNA helicases as some family members have been documented to be dysregulated in neurological and neurodevelopment disorders, as well as in cancers. Although different functional properties of RNA helicases offer multiple opportunities for small molecule development, molecular staples have recently come to the forefront. These bifunctional molecules interact with both protein and RNA components to lock them together, thereby imparting novel gain-of-function properties to their targets. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Small Molecule-RNA Interactions RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Sai Kiran Naineni
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Francis Robert
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Bhushan Nagar
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada.,Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Harris J, Borg NA. The multifaceted roles of NLRP3-modulating proteins in virus infection. Front Immunol 2022; 13:987453. [PMID: 36110852 PMCID: PMC9468583 DOI: 10.3389/fimmu.2022.987453] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022] Open
Abstract
The innate immune response to viruses is critical for the correct establishment of protective adaptive immunity. Amongst the many pathways involved, the NLRP3 [nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3)] inflammasome has received considerable attention, particularly in the context of immunity and pathogenesis during infection with influenza A (IAV) and SARS-CoV-2, the causative agent of COVID-19. Activation of the NLRP3 inflammasome results in the secretion of the proinflammatory cytokines IL-1β and IL-18, commonly coupled with pyroptotic cell death. While this mechanism is protective and key to host defense, aberrant NLRP3 inflammasome activation causes a hyperinflammatory response and excessive release of cytokines, both locally and systemically. Here, we discuss key molecules in the NLRP3 pathway that have also been shown to have significant roles in innate and adaptive immunity to viruses, including DEAD box helicase X-linked (DDX3X), vimentin and macrophage migration inhibitory factor (MIF). We also discuss the clinical opportunities to suppress NLRP3-mediated inflammation and reduce disease severity.
Collapse
Affiliation(s)
- James Harris
- Cell Biology Assays Team, Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Centre for Inflammatory diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Natalie A. Borg
- Immunity and Immune Evasion Laboratory, Chronic Infectious and Inflammatory Diseases Research, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
7
|
Brai A, Trivisani CI, Poggialini F, Pasqualini C, Vagaggini C, Dreassi E. DEAD-Box Helicase DDX3X as a Host Target against Emerging Viruses: New Insights for Medicinal Chemical Approaches. J Med Chem 2022; 65:10195-10216. [PMID: 35899912 DOI: 10.1021/acs.jmedchem.2c00755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In recent years, globalization, global warming, and population aging have contributed to the spread of emerging viruses, such as coronaviruses (COVs), West Nile (WNV), Dengue (DENV), and Zika (ZIKV). The number of reported infections is increasing, and considering the high viral mutation rate, it is conceivable that it will increase significantly in the coming years. The risk caused by viruses is now more evident due to the COVID-19 pandemic, which highlighted the need to find new broad-spectrum antiviral agents able to tackle the present pandemic and future epidemics. DDX3X helicase is a host factor required for viral replication. Selective inhibitors have been identified and developed into broad-spectrum antivirals active against emerging pathogens, including SARS-CoV-2 and most importantly against drug-resistant strains. This perspective describes the inhibitors identified in the last years, highlighting their therapeutic potential as innovative broad-spectrum antivirals.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnology, Chemistry & Pharmacy, University of Siena, I-53100 Siena Italy
| | | | - Federica Poggialini
- Department of Biotechnology, Chemistry & Pharmacy, University of Siena, I-53100 Siena Italy
| | - Claudia Pasqualini
- Department of Biotechnology, Chemistry & Pharmacy, University of Siena, I-53100 Siena Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry & Pharmacy, University of Siena, I-53100 Siena Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry & Pharmacy, University of Siena, I-53100 Siena Italy
| |
Collapse
|
8
|
Rao S, Mahmoudi T. DEAD-ly Affairs: The Roles of DEAD-Box Proteins on HIV-1 Viral RNA Metabolism. Front Cell Dev Biol 2022; 10:917599. [PMID: 35769258 PMCID: PMC9234453 DOI: 10.3389/fcell.2022.917599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
In order to ensure viral gene expression, Human Immunodeficiency virus type-1 (HIV-1) recruits numerous host proteins that promote optimal RNA metabolism of the HIV-1 viral RNAs (vRNAs), such as the proteins of the DEAD-box family. The DEAD-box family of RNA helicases regulates multiple steps of RNA metabolism and processing, including transcription, splicing, nucleocytoplasmic export, trafficking, translation and turnover, mediated by their ATP-dependent RNA unwinding ability. In this review, we provide an overview of the functions and role of all DEAD-box family protein members thus far described to influence various aspects of HIV-1 vRNA metabolism. We describe the molecular mechanisms by which HIV-1 hijacks these host proteins to promote its gene expression and we discuss the implications of these interactions during viral infection, their possible roles in the maintenance of viral latency and in inducing cell death. We also speculate on the emerging potential of pharmacological inhibitors of DEAD-box proteins as novel therapeutics to control the HIV-1 pandemic.
Collapse
Affiliation(s)
- Shringar Rao
- Department of Biochemistry, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Centre, Rotterdam, Netherlands
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, Netherlands
- Department of Urology, Erasmus University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
9
|
Recent research results have converted gp120 binders to a therapeutic option for the treatment of HIV-1 infection. A medicinal chemistry point of view. Eur J Med Chem 2021; 229:114078. [PMID: 34992041 DOI: 10.1016/j.ejmech.2021.114078] [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: 11/19/2021] [Revised: 12/16/2021] [Accepted: 12/22/2021] [Indexed: 11/21/2022]
Abstract
Current therapeutic armamentarium for treatment of HIV-1 infection is based on the use of highly active antiretroviral therapy that, unfortunately, does not act as a curative remedy. Moreover, duration of the therapy often results in lack of compliance with the consequent emergence of multidrug resistance. Finally, drug toxicity issues also arise during treatments. In the attempt to achieve a curative effect, in addition to invest substantial resources in finding new anti-HIV-1 agents and in optimizing antiviral lead compounds and drugs currently available, additional efforts should be done to deplete viral reservoir located within host CD4+ T cells. Gp120 binders represent a class of compounds able to affect the interactions between viral envelope proteins and host CD4, thus avoiding virus-to-cell attachment and fusion, and the consequent viral entry into host cells. This review summarizes the efforts done in the last five years to design new gp120 binders, that finally culminated in the approval of fostemsavir as an anti-HIV-1 drug.
Collapse
|
10
|
Gherardini L, Inzalaco G, Imperatore F, D’Aurizio R, Franci L, Miragliotta V, Boccuto A, Calandro P, Andreini M, Tarditi A, Chiariello M. The FHP01 DDX3X Helicase Inhibitor Exerts Potent Anti-Tumor Activity In Vivo in Breast Cancer Pre-Clinical Models. Cancers (Basel) 2021; 13:cancers13194830. [PMID: 34638314 PMCID: PMC8507746 DOI: 10.3390/cancers13194830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/25/2022] Open
Abstract
Inhibition of DDX3X expression or activity reduces proliferation in cells from various tumor tissues, in particular in breast cancer, and its expression often correlates to tumor aggressiveness. This makes DDX3X a prominent candidate for the design of drugs for novel personalized therapeutic strategies. Starting from an in silico drug discovery approach, a group of molecules has been selected by molecular docking at the RNA binding site of DDX3X. Here, the most promising among them, FHP01, was evaluated in breast cancer preclinical models. Specifically, FHP01 exhibited very effective antiproliferative and killing activity against different breast cancer cell types, among which those from triple-negative breast cancer (TNBC). Interestingly, FHP01 also inhibited WNT signaling, a key tumorigenic pathway already correlated to DDX3X functions in breast cancer model cell lines. Ultimately, FHP01 also caused a significant reduction, in vivo, in the growth of MDA MB 231-derived TNBC xenograft models. Importantly, FHP01 showed good bioavailability and no toxicity on normal peripheral blood mononuclear cells in vitro and on several mouse tissues in vivo. Overall, our data suggest that the use of FHP01 and its related compounds may represent a novel therapeutic approach with high potential against breast cancer, including the triple-negative subtype usually correlated to the most unfavorable outcomes because of the lack of available targeted therapies.
Collapse
Affiliation(s)
- Lisa Gherardini
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.G.); (G.I.); (F.I.); (L.F.)
| | - Giovanni Inzalaco
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.G.); (G.I.); (F.I.); (L.F.)
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy;
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Francesco Imperatore
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.G.); (G.I.); (F.I.); (L.F.)
| | - Romina D’Aurizio
- Istituto di Informatica e Telematica (IIT), Consiglio Nazionale delle Ricerche (CNR), 56124 Pisa, Italy;
| | - Lorenzo Franci
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.G.); (G.I.); (F.I.); (L.F.)
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy;
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | | | - Adele Boccuto
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Pierpaolo Calandro
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy;
| | - Matteo Andreini
- First Health Pharmaceutical B.V., 1098 XH Amsterdam, The Netherlands; (M.A.); (A.T.)
| | - Alessia Tarditi
- First Health Pharmaceutical B.V., 1098 XH Amsterdam, The Netherlands; (M.A.); (A.T.)
| | - Mario Chiariello
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.G.); (G.I.); (F.I.); (L.F.)
- Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy;
- Correspondence: ; Tel.: +39-057-723-1274
| |
Collapse
|
11
|
Arginine Methylation of hnRNPK Inhibits the DDX3-hnRNPK Interaction to Play an Anti-Apoptosis Role in Osteosarcoma Cells. Int J Mol Sci 2021; 22:ijms22189764. [PMID: 34575922 PMCID: PMC8469703 DOI: 10.3390/ijms22189764] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
Heterogeneous nuclear ribonucleoprotein K (hnRNPK) is an RNA/DNA binding protein involved in diverse cell processes; it is also a p53 coregulator that initiates apoptosis under DNA damage conditions. However, the upregulation of hnRNPK is correlated with cancer transformation, progression, and migration, whereas the regulatory role of hnRNPK in cancer malignancy remains unclear. We previously showed that arginine methylation of hnRNPK attenuated the apoptosis of U2OS osteosarcoma cells under DNA damage conditions, whereas the replacement of endogenous hnRNPK with a methylation-defective mutant inversely enhanced apoptosis. The present study further revealed that an RNA helicase, DDX3, whose C-terminus preferentially binds to the unmethylated hnRNPK and could promote such apoptotic enhancement. Moreover, C-terminus-truncated DDX3 induced significantly less apoptosis than full-length DDX3. Notably, we also identified a small molecule that docks at the ATP-binding site of DDX3, promotes the DDX3-hnRNPK interaction, and induces further apoptosis. Overall, we have shown that the arginine methylation of hnRNPK suppresses the apoptosis of U2OS cells via interfering with DDX3-hnRNPK interaction. On the other hand, DDX3-hnRNPK interaction with a proapoptotic role may serve as a target for promoting apoptosis in osteosarcoma cells.
Collapse
|
12
|
Sergeeva O, Abakumova T, Kurochkin I, Ialchina R, Kosyreva A, Prikazchikova T, Varlamova V, Shcherbinina E, Zatsepin T. Level of Murine DDX3 RNA Helicase Determines Phenotype Changes of Hepatocytes In Vitro and In Vivo. Int J Mol Sci 2021; 22:ijms22136958. [PMID: 34203429 PMCID: PMC8269429 DOI: 10.3390/ijms22136958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 11/26/2022] Open
Abstract
DDX3 RNA helicase is intensively studied as a therapeutic target due to participation in the replication of some viruses and involvement in cancer progression. Here we used transcriptome analysis to estimate the primary response of hepatocytes to different levels of RNAi-mediated knockdown of DDX3 RNA helicase both in vitro and in vivo. We found that a strong reduction of DDX3 protein (>85%) led to similar changes in vitro and in vivo—deregulation of the cell cycle and Wnt and cadherin pathways. Also, we observed the appearance of dead hepatocytes in the healthy liver and a decrease of cell viability in vitro after prolonged treatment. However, more modest downregulation of the DDX3 protein (60–65%) showed discordant results in vitro and in vivo—similar changes in vitro as in the case of strong knockdown and a different phenotype in vivo. These results demonstrate that the level of DDX3 protein can dramatically influence the cell phenotype in vivo and the decrease of DDX3, for more than 85% leads to cell death in normal tissues, which should be taken into account during the drug development of DDX3 inhibitors.
Collapse
Affiliation(s)
- Olga Sergeeva
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
- Correspondence: ; Tel.: +7-926-388-0865
| | - Tatiana Abakumova
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
| | - Ilia Kurochkin
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
| | - Renata Ialchina
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
| | - Anna Kosyreva
- Research Institute of Human Morphology, 117418 Moscow, Russia;
| | - Tatiana Prikazchikova
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
| | - Varvara Varlamova
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
| | - Evgeniya Shcherbinina
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
| | - Timofei Zatsepin
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia; (T.A.); (I.K.); (R.I.); (T.P.); (V.V.); (E.S.); (T.Z.)
- Department of Chemistry, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
13
|
RNA Helicase DDX3: A Double-Edged Sword for Viral Replication and Immune Signaling. Microorganisms 2021; 9:microorganisms9061206. [PMID: 34204859 PMCID: PMC8227550 DOI: 10.3390/microorganisms9061206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022] Open
Abstract
DDX3 is a cellular ATP-dependent RNA helicase involved in different aspects of RNA metabolism ranging from transcription to translation and therefore, DDX3 participates in the regulation of key cellular processes including cell cycle progression, apoptosis, cancer and the antiviral immune response leading to type-I interferon production. DDX3 has also been described as an essential cellular factor for the replication of different viruses, including important human threats such HIV-1 or HCV, and different small molecules targeting DDX3 activity have been developed. Indeed, increasing evidence suggests that DDX3 can be considered not only a promising but also a viable target for anticancer and antiviral treatments. In this review, we summarize distinct functional aspects of DDX3 focusing on its participation as a double-edged sword in the host immune response and in the replication cycle of different viruses.
Collapse
|
14
|
Selective cell death in HIV-1-infected cells by DDX3 inhibitors leads to depletion of the inducible reservoir. Nat Commun 2021; 12:2475. [PMID: 33931637 PMCID: PMC8087668 DOI: 10.1038/s41467-021-22608-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/15/2021] [Indexed: 02/02/2023] Open
Abstract
An innovative approach to eliminate HIV-1-infected cells emerging out of latency, the major hurdle to HIV-1 cure, is to pharmacologically reactivate viral expression and concomitantly trigger intracellular pro-apoptotic pathways in order to selectively induce cell death (ICD) of infected cells, without reliance on the extracellular immune system. In this work, we demonstrate the effect of DDX3 inhibitors on selectively inducing cell death in latent HIV-1-infected cell lines, primary CD4+ T cells and in CD4+ T cells from cART-suppressed people living with HIV-1 (PLWHIV). We used single-cell FISH-Flow technology to characterise the contribution of viral RNA to inducing cell death. The pharmacological targeting of DDX3 induced HIV-1 RNA expression, resulting in phosphorylation of IRF3 and upregulation of IFNβ. DDX3 inhibition also resulted in the downregulation of BIRC5, critical to cell survival during HIV-1 infection, and selectively induced apoptosis in viral RNA-expressing CD4+ T cells but not bystander cells. DDX3 inhibitor treatment of CD4+ T cells from PLWHIV resulted in an approximately 50% reduction of the inducible latent HIV-1 reservoir by quantitation of HIV-1 RNA, by FISH-Flow, RT-qPCR and TILDA. This study provides proof of concept for pharmacological reversal of latency coupled to induction of apoptosis towards the elimination of the inducible reservoir.
Collapse
|
15
|
Putra V, Hulme AJ, Tee AE, Sun JQ, Atmadibrata B, Ho N, Chen J, Gao J, Norris MD, Haber M, Kavallaris M, Henderson MJ, McCarroll J, Trahair T, Liu T, Liu PY. The RNA-helicase DDX21 upregulates CEP55 expression and promotes neuroblastoma. Mol Oncol 2021; 15:1162-1179. [PMID: 33497018 PMCID: PMC8024731 DOI: 10.1002/1878-0261.12906] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Approximately 25% of human neuroblastoma is caused by amplification of the MYCN oncogene, which leads to overexpression of N-Myc oncoprotein. The survival rate for this patient subtype is <50%. Here, we show that N-Myc protein bound to the DEAD-box RNA helicase DDX21 gene promoter and upregulated DDX21 mRNA and protein expression. Genome-wide differential gene expression studies identified centrosomal protein CEP55 as one of the genes most dramatically downregulated after DDX21 knockdown in MYCN-amplified neuroblastoma cells. Knocking down DDX21 or CEP55 reduced neuroblastoma cell cytoskeleton stability and cell proliferation and all but abolished clonogenic capacity. Importantly, DDX21 knockdown initially induced tumor regression in neuroblastoma-bearing mice and suppressed tumor progression. In human neuroblastoma tissues, a high level of DDX21 expression correlated with a high level of N-Myc expression and with CEP55 expression, and independently predicted poor patient prognosis. Taken together, our data show that DDX21 induces CEP55 expression, MYCN-amplified neuroblastoma cell proliferation, and tumorigenesis, and that DDX21 and CEP55 are valid therapeutic targets for the treatment of MYCN-amplified neuroblastoma.
Collapse
Affiliation(s)
- Vina Putra
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Amy J. Hulme
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Andrew E. Tee
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Jane Q.J. Sun
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Bernard Atmadibrata
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Nicholas Ho
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Jingwei Chen
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Jixuan Gao
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Murray D. Norris
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
- University of New South Wales Centre for Childhood Cancer ResearchSydneyNSWAustralia
| | - Michelle Haber
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Maria Kavallaris
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and TechnologyAustralian Centre for NanomedicineUNSW SydneyKensingtonNSWAustralia
- School of Women’s and Children’s HealthFaculty of MedicineUNSW SydneyKensingtonNSWAustralia
| | - Michelle J. Henderson
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Joshua McCarroll
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Toby Trahair
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Tao Liu
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| | - Pei Y. Liu
- Children’s Cancer InstituteLowy Cancer Research CentreUNSW SydneyKensingtonNSWAustralia
| |
Collapse
|
16
|
Ali MAM. DEAD-box RNA helicases: The driving forces behind RNA metabolism at the crossroad of viral replication and antiviral innate immunity. Virus Res 2021; 296:198352. [PMID: 33640359 DOI: 10.1016/j.virusres.2021.198352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023]
Abstract
DEAD-box RNA helicases, the largest family of superfamily 2 helicases, are a profoundly conserved family of RNA-binding proteins, containing a distinctive Asp-Glu-Ala-Asp (D-E-A-D) sequence motif, which is the origin of their name. Aside from the ATP-dependent unwinding of RNA duplexes, which set up these proteins as RNA helicases, DEAD-box proteins have been found to additionally stimulate RNA duplex fashioning and to uproot proteins from RNA, aiding the reformation of RNA and RNA-protein complexes. There is accumulating evidence that DEAD-box helicases play functions in the recognition of foreign nucleic acids and the modification of viral infection. As intracellular parasites, viruses must avoid identification by innate immune sensing mechanisms and disintegration by cellular machinery, whilst additionally exploiting host cell activities to assist replication. The capability of DEAD-box helicases to sense RNA in a sequence-independent way, as well as the broadness of cellular roles performed by members of this family, drive them to affect innate sensing and viral infections in numerous manners. Undoubtedly, DEAD-box helicases have been demonstrated to contribute to intracellular immune recognition, function as antiviral effectors, and even to be exploited by viruses to support their replication. Relying on the virus or the viral cycle phase, a DEAD-box helicase can function either in a proviral manner or as an antiviral factor. This review gives a comprehensive perspective on the various biochemical characteristics of DEAD-box helicases and their links to structural data. It additionally outlines the multiple functions that members of the DEAD-box helicase family play during viral infections.
Collapse
Affiliation(s)
- Mohamed A M Ali
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| |
Collapse
|
17
|
Abdelkrim YZ, Banroques J, Kyle Tanner N. Known Inhibitors of RNA Helicases and Their Therapeutic Potential. Methods Mol Biol 2021; 2209:35-52. [PMID: 33201461 DOI: 10.1007/978-1-0716-0935-4_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RNA helicases are proteins found in all kingdoms of life, and they are associated with all processes involving RNA from transcription to decay. They use NTP binding and hydrolysis to unwind duplexes, to remodel RNA structures and protein-RNA complexes, and to facilitate the unidirectional metabolism of biological processes. Viral, bacterial, and eukaryotic parasites have an intimate need for RNA helicases in their reproduction. Moreover, various disorders, like cancers, are often associated with a perturbation of the host's helicase activity. Thus, RNA helicases provide a rich source of targets for the development of therapeutic or prophylactic drugs. In this review, we provide an overview of the different targeting strategies against helicases, the different types of compounds explored, the proposed inhibitory mechanisms of the compounds on the proteins, and the therapeutic potential of these compounds in the treatment of various disorders.
Collapse
Affiliation(s)
- Yosser Zina Abdelkrim
- Expression Génétique Microbienne, UMR8261 CNRS, Institut de Biologie Physico-Chimique, Université de Paris, Paris, France.,Molecular Epidemiology and Experimental Pathology (LR16IPT04), Institut Pasteur de Tunis/Université de Tunis el Manar, Tunis-Belvédère, Tunisia
| | - Josette Banroques
- Expression Génétique Microbienne, UMR8261 CNRS, Institut de Biologie Physico-Chimique, Université de Paris, Paris, France.,PSL Research University, Paris, France
| | - N Kyle Tanner
- Expression Génétique Microbienne, UMR8261 CNRS, Institut de Biologie Physico-Chimique, Université de Paris, Paris, France.
| |
Collapse
|
18
|
Felicetti T, Manfroni G, Cecchetti V, Cannalire R. Broad-Spectrum Flavivirus Inhibitors: a Medicinal Chemistry Point of View. ChemMedChem 2020; 15:2391-2419. [PMID: 32961008 DOI: 10.1002/cmdc.202000464] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/16/2020] [Indexed: 12/16/2022]
Abstract
Infections by flaviviruses, such as Dengue, West Nile, Yellow Fever and Zika viruses, represent a growing risk for global health. There are vaccines only for few flaviviruses while no effective treatments are available. Flaviviruses share epidemiological, structural, and ecologic features and often different viruses can co-infect the same host. Therefore, the identification of broad-spectrum inhibitors is highly desirable either for known flaviviruses or for viruses that likely will emerge in the future. Strategies targeting both virus and host factors have been pursued to identify broad-spectrum antiflaviviral agents. In this review, we describe the most promising and best characterized targets and their relative broad-spectrum inhibitors, identified by drug repurposing/libraries screenings and by focused medicinal chemistry campaigns. Finally, we discuss about future strategies to identify new broad-spectrum antiflavivirus agents.
Collapse
Affiliation(s)
- Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Rolando Cannalire
- Department of Pharmacy, University of Napoli "Federico II", via D. Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
19
|
Weiße J, Rosemann J, Krauspe V, Kappler M, Eckert AW, Haemmerle M, Gutschner T. RNA-Binding Proteins as Regulators of Migration, Invasion and Metastasis in Oral Squamous Cell Carcinoma. Int J Mol Sci 2020; 21:E6835. [PMID: 32957697 PMCID: PMC7555251 DOI: 10.3390/ijms21186835] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Nearly 7.5% of all human protein-coding genes have been assigned to the class of RNA-binding proteins (RBPs), and over the past decade, RBPs have been increasingly recognized as important regulators of molecular and cellular homeostasis. RBPs regulate the post-transcriptional processing of their target RNAs, i.e., alternative splicing, polyadenylation, stability and turnover, localization, or translation as well as editing and chemical modification, thereby tuning gene expression programs of diverse cellular processes such as cell survival and malignant spread. Importantly, metastases are the major cause of cancer-associated deaths in general, and particularly in oral cancers, which account for 2% of the global cancer mortality. However, the roles and architecture of RBPs and RBP-controlled expression networks during the diverse steps of the metastatic cascade are only incompletely understood. In this review, we will offer a brief overview about RBPs and their general contribution to post-transcriptional regulation of gene expression. Subsequently, we will highlight selected examples of RBPs that have been shown to play a role in oral cancer cell migration, invasion, and metastasis. Last but not least, we will present targeting strategies that have been developed to interfere with the function of some of these RBPs.
Collapse
Affiliation(s)
- Jonas Weiße
- Junior Research Group ‘RNA Biology and Pathogenesis’, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (J.W.); (J.R.); (V.K.)
| | - Julia Rosemann
- Junior Research Group ‘RNA Biology and Pathogenesis’, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (J.W.); (J.R.); (V.K.)
| | - Vanessa Krauspe
- Junior Research Group ‘RNA Biology and Pathogenesis’, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (J.W.); (J.R.); (V.K.)
| | - Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Alexander W. Eckert
- Department of Cranio Maxillofacial Surgery, Paracelsus Medical University, 90471 Nuremberg, Germany;
| | - Monika Haemmerle
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany;
| | - Tony Gutschner
- Junior Research Group ‘RNA Biology and Pathogenesis’, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany; (J.W.); (J.R.); (V.K.)
| |
Collapse
|
20
|
Dash RN, Moharana AK, Subudhi BB. Sulfonamides: Antiviral Strategy for Neglected Tropical Disease Virus. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200515094100] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The viral infections are a threat to the health system around the globe. Although
more than 60 antiviral drugs have been approved by the FDA, most of them are for the
management of few viruses like HIV, Hepatitis and Influenza. There is no antiviral for
many viruses including Dengue, Chikungunya and Japanese encephalitis. Many of these
neglected viruses are increasingly becoming global pathogens. Lack of broad spectrum of
action and the rapid rise of resistance and cross-resistance to existing antiviral have further
increased the challenge of antiviral development. Sulfonamide, as a privileged scaffold,
has been capitalized to develop several bioactive compounds and drugs. Accordingly, several
reviews have been published in recent times on bioactive sulfonamides. However,
there are not enough review reports of antiviral sulfonamides in the last five years. Sulfonamides
scaffolds have received sufficient attention for the development of non- nucleoside antivirals following
the emergence of cross-resistance to nucleoside inhibitors. Hybridization of bioactive pharmacophores
with sulfonamides has been used as a strategy to develop sulfonamide antivirals. This review is an effort to
analyze these attempts and evaluate their translational potential. Parameters including potency (IC50), toxicity
(CC50) and selectivity (CC50/IC50) have been used in this report to suggest the potential of sulfonamide derivatives
to progress further as antiviral. Since most of these antiviral properties are based on the in vitro results,
the drug-likeness of molecules has been predicted to propose in vivo potential. The structure-activity relationship
has been analyzed to encourage further optimization of antiviral properties.
Collapse
Affiliation(s)
- Rudra Narayan Dash
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751029, Odisha, India
| | - Alok Kumar Moharana
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751029, Odisha, India
| | - Bharat Bhusan Subudhi
- Drug Development and Analysis Laboratory, School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751029, Odisha, India
| |
Collapse
|
21
|
A Computational Approach with Biological Evaluation: Combinatorial Treatment of Curcumin and Exemestane Synergistically Regulates DDX3 Expression in Cancer Cell Lines. Biomolecules 2020; 10:biom10060857. [PMID: 32512851 PMCID: PMC7355417 DOI: 10.3390/biom10060857] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 01/07/2023] Open
Abstract
DDX3 belongs to RNA helicase family that demonstrates oncogenic properties and has gained wider attention due to its role in cancer progression, proliferation and transformation. Mounting reports have evidenced the role of DDX3 in cancers making it a promising target to abrogate DDX3 triggered cancers. Dual pharmacophore models were generated and were subsequently validated. They were used as 3D queries to screen the InterBioScreen database, resulting in the selection of curcumin that was escalated to molecular dynamics simulation studies. In vitro anti-cancer analysis was conducted on three cell lines such as MCF-7, MDA-MB-231 and HeLa, which were evaluated along with exemestane. Curcumin was docked into the active site of the protein target (PDB code 2I4I) to estimate the binding affinity. The compound has interacted with two key residues and has displayed stable molecular dynamics simulation results. In vitro analysis has demonstrated that both the candidate compounds have reduced the expression of DDX3 in three cell lines. However, upon combinatorial treatment of curcumin (10 and 20 μM) and exemestane (50 μM) a synergism was exhibited, strikingly downregulating the DDX3 expression and has enhanced apoptosis in three cell lines. The obtained results illuminate the use of curcumin as an alternative DDX3 inhibitor and can serve as a chemical scaffold to design new small molecules.
Collapse
|
22
|
Quaranta P, Lottini G, Chesi G, Contrafatto F, Russotto R, Macera L, Lai M, Spezia PG, Brai A, Botta M, Freer G, Pistello M. DDX3 inhibitors show antiviral activity against positive-sense single-stranded RNA viruses but not against negative-sense single-stranded RNA viruses: The coxsackie B model. Antiviral Res 2020; 178:104750. [DOI: 10.1016/j.antiviral.2020.104750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 02/08/2020] [Accepted: 02/18/2020] [Indexed: 10/24/2022]
|
23
|
Brai A, Boccuto A, Monti M, Marchi S, Vicenti I, Saladini F, Trivisani CI, Pollutri A, Trombetta CM, Montomoli E, Riva V, Garbelli A, Nola EM, Zazzi M, Maga G, Dreassi E, Botta M. Exploring the Implication of DDX3X in DENV Infection: Discovery of the First-in-Class DDX3X Fluorescent Inhibitor. ACS Med Chem Lett 2020; 11:956-962. [PMID: 32435411 DOI: 10.1021/acsmedchemlett.9b00681] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
In the absence of effective drugs or vaccines for the treatment of the five Dengue Virus serotypes, the search for novel antiviral drugs is of primary importance for the scientific community. In this context, drug repurposing represents the most used strategy; however, the study of host targets is now attracting attention since it allows identification of broad-spectrum drugs endowed with high genetic barrier. In the last ten years our research group identified several small molecules DDX3X inhibitors and proved their efficacy against different viruses including novel emerging ones. Herein, starting from a screening of our compounds, we designed and synthesized novel derivatives with potent activity and high selectivity. Finally, we synthesized a fluorescent inhibitor that allowed us to study DDX3X cellular localization during DENV infection in vitro. Immunofluorescence analysis showed that our inhibitor colocalized with DDX3X, promoting the reduction of infected cells and recovering the number of viable cells.
Collapse
Affiliation(s)
- Annalaura Brai
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Adele Boccuto
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, 53100 Siena, Italy
| | - Martina Monti
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università degli Studi di Siena, 53100 Siena, Italy
| | - Serena Marchi
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università degli Studi di Siena, 53100 Siena, Italy
| | - Ilaria Vicenti
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, 53100 Siena, Italy
| | - Francesco Saladini
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, 53100 Siena, Italy
| | | | - Alessandro Pollutri
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Claudia Maria Trombetta
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università degli Studi di Siena, 53100 Siena, Italy
| | - Emanuele Montomoli
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università degli Studi di Siena, 53100 Siena, Italy
- VisMederi Srl, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Valentina Riva
- Institute of Molecular Genetics IGM-CNR “Luigi Luca Cavalli-Sforza”, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Anna Garbelli
- Institute of Molecular Genetics IGM-CNR “Luigi Luca Cavalli-Sforza”, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Emanuele Maria Nola
- Institute of Molecular Genetics IGM-CNR “Luigi Luca Cavalli-Sforza”, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Maurizio Zazzi
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, 53100 Siena, Italy
| | - Giovanni Maga
- Institute of Molecular Genetics IGM-CNR “Luigi Luca Cavalli-Sforza”, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Elena Dreassi
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Maurizio Botta
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, via Aldo Moro 2, 53100 Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, BioLife Science Building, Suite 333, 1900 N 12th Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
24
|
Brai A, Riva V, Saladini F, Zamperini C, Trivisani CI, Garbelli A, Pennisi C, Giannini A, Boccuto A, Bugli F, Martini M, Sanguinetti M, Zazzi M, Dreassi E, Botta M, Maga G. DDX3X inhibitors, an effective way to overcome HIV-1 resistance targeting host proteins. Eur J Med Chem 2020; 200:112319. [PMID: 32446036 DOI: 10.1016/j.ejmech.2020.112319] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
The huge resources that had gone into Human Immunodeficiency virus (HIV) research led to the development of potent antivirals able to suppress viral load in the majority of treated patients, thus dramatically increasing the life expectancy of people living with HIV. However, life-long treatments could result in the emergence of drug-resistant viruses that can progressively reduce the number of therapeutic options, facilitating the progression of the disease. In this scenario, we previously demonstrated that inhibitors of the human DDX3X helicase can represent an innovative approach for the simultaneous treatment of HIV and other viral infections such as Hepatitis c virus (HCV). We reported herein 6b, a novel DDX3X inhibitor that thanks to its distinct target of action is effective against HIV-1 strains resistant to currently approved drugs. Its improved in vitro ADME properties allowed us to perform preliminary in vivo studies in mice, which highlighted optimal biocompatibility and an improved bioavailability. These results represent a significant advancement in the development of DDX3X inhibitors as a novel class of broad spectrum and safe anti-HIV-1 drugs.
Collapse
Affiliation(s)
- Annalaura Brai
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. De Gasperi 2, I-53100, Siena, Italy
| | - Valentina Riva
- Istituto di Genetica Molecolare "Luigi Luca Cavalli - Sforza", IGM-CNR, Via Abbiategrasso 207, I-27100, Pavia, Italy
| | - Francesco Saladini
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, I-53100, Siena, Italy
| | - Claudio Zamperini
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. De Gasperi 2, I-53100, Siena, Italy
| | - Claudia Immacolata Trivisani
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. De Gasperi 2, I-53100, Siena, Italy
| | - Anna Garbelli
- Istituto di Genetica Molecolare "Luigi Luca Cavalli - Sforza", IGM-CNR, Via Abbiategrasso 207, I-27100, Pavia, Italy
| | - Carla Pennisi
- Istituto di Genetica Molecolare "Luigi Luca Cavalli - Sforza", IGM-CNR, Via Abbiategrasso 207, I-27100, Pavia, Italy
| | - Alessia Giannini
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, I-53100, Siena, Italy
| | - Adele Boccuto
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, I-53100, Siena, Italy
| | - Francesca Bugli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, I-00168, Roma, Italy; Istituto di Microbiologia, Università Cattolica del SC, L.go F. Vito 1, I-00168, Roma, Italy
| | - Maurizio Martini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Servizio di Istopatologia e Citodiagnosi, Rome, Italy; Istituto di Patologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Sanguinetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, I-00168, Roma, Italy; Istituto di Microbiologia, Università Cattolica del SC, L.go F. Vito 1, I-00168, Roma, Italy
| | - Maurizio Zazzi
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, I-53100, Siena, Italy
| | - Elena Dreassi
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. De Gasperi 2, I-53100, Siena, Italy
| | - Maurizio Botta
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. De Gasperi 2, I-53100, Siena, Italy; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, BioLife Science Building, Suite 333, 1900 N 12th Street, Philadelphia, PA, 19122, USA.
| | - Giovanni Maga
- Istituto di Genetica Molecolare "Luigi Luca Cavalli - Sforza", IGM-CNR, Via Abbiategrasso 207, I-27100, Pavia, Italy.
| |
Collapse
|
25
|
Brosh RM, Matson SW. History of DNA Helicases. Genes (Basel) 2020; 11:genes11030255. [PMID: 32120966 PMCID: PMC7140857 DOI: 10.3390/genes11030255] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Since the discovery of the DNA double helix, there has been a fascination in understanding the molecular mechanisms and cellular processes that account for: (i) the transmission of genetic information from one generation to the next and (ii) the remarkable stability of the genome. Nucleic acid biologists have endeavored to unravel the mysteries of DNA not only to understand the processes of DNA replication, repair, recombination, and transcription but to also characterize the underlying basis of genetic diseases characterized by chromosomal instability. Perhaps unexpectedly at first, DNA helicases have arisen as a key class of enzymes to study in this latter capacity. From the first discovery of ATP-dependent DNA unwinding enzymes in the mid 1970's to the burgeoning of helicase-dependent pathways found to be prevalent in all kingdoms of life, the story of scientific discovery in helicase research is rich and informative. Over four decades after their discovery, we take this opportunity to provide a history of DNA helicases. No doubt, many chapters are left to be written. Nonetheless, at this juncture we are privileged to share our perspective on the DNA helicase field - where it has been, its current state, and where it is headed.
Collapse
Affiliation(s)
- Robert M. Brosh
- Section on DNA Helicases, Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Correspondence: (R.M.B.J.); (S.W.M.); Tel.: +1-410-558-8578 (R.M.B.J.); +1-919-962-0005 (S.W.M.)
| | - Steven W. Matson
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: (R.M.B.J.); (S.W.M.); Tel.: +1-410-558-8578 (R.M.B.J.); +1-919-962-0005 (S.W.M.)
| |
Collapse
|
26
|
DEAD-box RNA Helicase DDX3: Functional Properties and Development of DDX3 Inhibitors as Antiviral and Anticancer Drugs. Molecules 2020; 25:molecules25041015. [PMID: 32102413 PMCID: PMC7070539 DOI: 10.3390/molecules25041015] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/05/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
This short review is focused on enzymatic properties of human ATP-dependent RNA helicase DDX3 and the development of antiviral and anticancer drugs targeting cellular helicases. DDX3 belongs to the DEAD-box proteins, a large family of RNA helicases that participate in all aspects of cellular processes, such as cell cycle progression, apoptosis, innate immune response, viral replication, and tumorigenesis. DDX3 has a variety of functions in the life cycle of different viruses. DDX3 helicase is required to facilitate both the Rev-mediated export of unspliced/partially spliced human immunodeficiency virus (HIV) RNA from nucleus and Tat-dependent translation of viral genes. DDX3 silencing blocks the replication of HIV, HCV, and some other viruses. On the other hand, DDX displays antiviral effect against Dengue virus and hepatitis B virus through the stimulation of interferon beta production. The role of DDX3 in different types of cancer is rather controversial. DDX3 acts as an oncogene in one type of cancer, but demonstrates tumor suppressor properties in other types. The human DDX3 helicase is now considered as a new attractive target for the development of novel pharmaceutical drugs. The most interesting inhibitors of DDX3 helicase and the mechanisms of their actions as antiviral or anticancer drugs are discussed in this short review.
Collapse
|
27
|
Ji X, Li Z. Medicinal chemistry strategies toward host targeting antiviral agents. Med Res Rev 2020; 40:1519-1557. [PMID: 32060956 PMCID: PMC7228277 DOI: 10.1002/med.21664] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 12/11/2022]
Abstract
Direct‐acting antiviral agents (DAAs) represent a class of drugs targeting viral proteins and have been demonstrated to be very successful in combating viral infections in clinic. However, DAAs suffer from several inherent limitations, including narrow‐spectrum antiviral profiles and liability to drug resistance, and hence there are still unmet needs in the treatment of viral infections. In comparison, host targeting antivirals (HTAs) target host factors for antiviral treatment. Since host proteins are probably broadly required for various viral infections, HTAs are not only perceived, but also demonstrated to exhibit broad‐spectrum antiviral activities. In addition, host proteins are not under the genetic control of viral genome, and hence HTAs possess much higher genetic barrier to drug resistance as compared with DAAs. In recent years, much progress has been made to the development of HTAs with the approval of chemokine receptor type 5 antagonist maraviroc for human immunodeficiency virus treatment and more in the pipeline for other viral infections. In this review, we summarize various host proteins as antiviral targets from a medicinal chemistry prospective. Challenges and issues associated with HTAs are also discussed.
Collapse
Affiliation(s)
- Xingyue Ji
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.,Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Lin TC. DDX3X Multifunctionally Modulates Tumor Progression and Serves as a Prognostic Indicator to Predict Cancer Outcomes. Int J Mol Sci 2019; 21:ijms21010281. [PMID: 31906196 PMCID: PMC6982152 DOI: 10.3390/ijms21010281] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/22/2022] Open
Abstract
DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, X-Linked (DDX3X), also known as DDX3, is one of the most widely studied and evolutionarily conserved members of the DEAD-box RNA helicase subfamily, and has been reported to participate in several cytosolic steps of mRNA metabolism. DDX3X facilitates the translation of specific targets via its helicase activity and regulates factors of the translation initiation complex. Emerging evidence illustrates the biological activities of DDX3X beyond its originally identified functions. The nonconventional regulatory effects include acting as a signaling adaptor molecule independent of enzymatic RNA remodeling, and DDX3X exhibits abnormal expression in cancers. DDX3X interacts with specific components to perform both oncogenic and tumor-suppressive roles in modulating tumor proliferation, migration, invasion, drug resistance, and cancer stemness in many types of cancers, indicating the need to unravel the associated molecular mechanisms. In this review article, we summarized and integrated current findings relevant to DDX3X in cancer research fields, cytokines and compounds modulating DDX3X's functions, and the released transcriptomic information and cancer patient clinical data from public databases. We found evidence for DDX3X having multiple impacts on cancer progression, and evaluated DDX3X expression levels in a pancancer panel and its associations with patient survival in each cancer-type cohort.
Collapse
Affiliation(s)
- Tsung-Chieh Lin
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
29
|
Synthesis and Antiviral Activity of Novel 1,3,4-Thiadiazole Inhibitors of DDX3X. Molecules 2019; 24:molecules24213988. [PMID: 31690062 PMCID: PMC6864647 DOI: 10.3390/molecules24213988] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/04/2019] [Accepted: 10/04/2019] [Indexed: 01/19/2023] Open
Abstract
The human ATPase/RNA helicase X-linked DEAD-box polypeptide 3 (DDX3X) emerged as a novel therapeutic target in the fight against both infectious diseases and cancer. Herein, a new family of DDX3X inhibitors was designed, synthesized, and tested for its inhibitory action on the ATPase activity of the enzyme. The potential use of the most promising derivatives it has been investigated by evaluating their anti-HIV-1 effects, revealing inhibitory activities in the low micromolar range. A preliminary ADME analysis demonstrated high metabolic stability and good aqueous solubility. The promising biological profile, together with the suitable in vitro pharmacokinetic properties, make these novel compounds a very good starting point for further development.
Collapse
|
30
|
Poynter SJ, Herrington-Krause S, DeWitte-Orr SJ. Two DExD/H-box helicases, DDX3 and DHX9, identified in rainbow trout are able to bind dsRNA. FISH & SHELLFISH IMMUNOLOGY 2019; 93:1056-1066. [PMID: 31340170 DOI: 10.1016/j.fsi.2019.07.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 06/10/2023]
Abstract
In mammals, the multifunctional DExH/D-box helicases, DDX3 and DHX9, are nucleic acid sensors with a role in antiviral immunity; their role in innate immunity in fish is not yet understood. In the present study, full-length DDX3 and DHX9 coding sequences were identified in rainbow trout (Oncorhynchus mykiss). Bioinformatic analysis demonstrated both deduced proteins were similar to those of other species, with ~80% identity to other fish species and ~70-75% identity to mammals, and both protein sequences had conserved domains found amongst all species. Phylogenetic analysis revealed clustering of DDX3 and DHX9 with corresponding proteins from other fish. Cellular localization of overexpressed DDX3 and DHX9 was performed using GFP-tagged proteins, and endogenous DDX3 localization was measured using immunocytochemistry. In the rainbow trout gonadal cell line, RTG-2, DHX9 localized mostly to the nucleus, while DDX3 was found mainly in the cytoplasm. Tissue distribution from healthy juvenile rainbow trout revealed ubiquitous constitutive expression, highest levels of DDX3 expression were seen in the liver and DHX9 levels were fairly consistent among all tissues tested. Stimulation of RTG-2 cells revealed that DDX3 and DHX9 transcripts were both significantly upregulated by treatment with the dsRNA molecule, poly I:C. A pull-down assay suggested both proteins were able to bind dsRNA. In addition to their roles in RNA metabolism, the conserved common domains found between the rainbow trout proteins and other species having defined antiviral roles, combined with the ability for the proteins to bind to dsRNA, suggest these proteins may play an important role in fish innate antiviral immunity. Future studies on both DDX3 and DHX9 function will contribute to a better understanding of teleost immunity.
Collapse
Affiliation(s)
- Sarah J Poynter
- Department of Biology, University of Waterloo, 200 University Ave W, Waterloo, ON, N2L 3G1, Canada
| | - Shanee Herrington-Krause
- Department of Biology, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - Stephanie J DeWitte-Orr
- Department of Biology, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada; Department of Health Sciences, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada.
| |
Collapse
|
31
|
Mohibi S, Chen X, Zhang J. Cancer the'RBP'eutics-RNA-binding proteins as therapeutic targets for cancer. Pharmacol Ther 2019; 203:107390. [PMID: 31302171 DOI: 10.1016/j.pharmthera.2019.07.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
Abstract
RNA-binding proteins (RBPs) play a critical role in the regulation of various RNA processes, including splicing, cleavage and polyadenylation, transport, translation and degradation of coding RNAs, non-coding RNAs and microRNAs. Recent studies indicate that RBPs not only play an instrumental role in normal cellular processes but have also emerged as major players in the development and spread of cancer. Herein, we review the current knowledge about RNA binding proteins and their role in tumorigenesis as well as the potential to target RBPs for cancer therapeutics.
Collapse
Affiliation(s)
- Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, United States
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, United States
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, United States.
| |
Collapse
|
32
|
From the magic bullet to the magic target: exploiting the diverse roles of DDX3X in viral infections and tumorigenesis. Future Med Chem 2019; 11:1357-1381. [PMID: 30816053 DOI: 10.4155/fmc-2018-0451] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
DDX3X is an ATPase/RNA helicase of the DEAD-box family and one of the most multifaceted helicases known up to date, acting in RNA metabolism, cell cycle control, apoptosis, stress response and innate immunity. Depending on the virus or the viral cycle stage, DDX3X can act either in a proviral fashion or as an antiviral factor. Similarly, in different cancer types, it can act either as an oncogene or a tumor-suppressor gene. Accumulating evidence indicated that DDX3X can be considered a promising target for anticancer and antiviral chemotherapy, but also that its exploitation requires a deeper understanding of the molecular mechanisms underlying its dual role in cancer and viral infections. In this Review, we will summarize the known roles of DDX3X in different tumor types and viral infections, and the different inhibitors available, illustrating the possible advantages and potential caveats of their use as anticancer and antiviral drugs.
Collapse
|
33
|
Brai A, Martelli F, Riva V, Garbelli A, Fazi R, Zamperini C, Pollutri A, Falsitta L, Ronzini S, Maccari L, Maga G, Giannecchini S, Botta M. DDX3X Helicase Inhibitors as a New Strategy To Fight the West Nile Virus Infection. J Med Chem 2019; 62:2333-2347. [PMID: 30721061 DOI: 10.1021/acs.jmedchem.8b01403] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Increased frequency of arbovirus outbreaks in the last 10 years represents an important emergence for global health. Climate warming, extensive urbanization of tropical regions, and human migration flows facilitate the expansion of anthropophilic mosquitos and the emerging or re-emerging of new viral infections. Only recently the human adenosinetriphosphatase/RNA helicase X-linked DEAD-box polypeptide 3 (DDX3X) emerged as a novel therapeutic target in the fight against infectious diseases. Herein, starting from our previous studies, a new family of DDX3X inhibitors was designed, synthesized, validated on the target enzyme, and evaluated against the West Nile virus (WNV) infection. Time of addition experiments after virus infection indicated that the compounds exerted their antiviral activities after the entry process, likely at the protein translation step of WNV replication. Finally, the most interesting compounds were then analyzed for their in vitro pharmacokinetic parameters, revealing favorable absorption, distribution, metabolism, and excretion values. The good safety profile together with a good activity against WNV for which no treatments are currently available, make this new class of molecules a good starting point for further in vivo studies.
Collapse
Affiliation(s)
- Annalaura Brai
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy.,Lead Discovery Siena S.r.l. , Castelnuovo Berardenga, I-53019 Siena , Italy
| | - Francesco Martelli
- Department of Experimental and Clinical Medicine , University of Florence , I-50134 Florence , Italy
| | - Valentina Riva
- Istituto di Genetica Molecolare, IGM-CNR , Via Abbiategrasso 207 , I-27100 Pavia , Italy
| | - Anna Garbelli
- Istituto di Genetica Molecolare, IGM-CNR , Via Abbiategrasso 207 , I-27100 Pavia , Italy
| | - Roberta Fazi
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy
| | - Claudio Zamperini
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy.,Lead Discovery Siena S.r.l. , Castelnuovo Berardenga, I-53019 Siena , Italy
| | - Alessandro Pollutri
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy
| | - Lucia Falsitta
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy
| | - Stefania Ronzini
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy
| | - Laura Maccari
- Lead Discovery Siena S.r.l. , Castelnuovo Berardenga, I-53019 Siena , Italy
| | - Giovanni Maga
- Istituto di Genetica Molecolare, IGM-CNR , Via Abbiategrasso 207 , I-27100 Pavia , Italy
| | - Simone Giannecchini
- Department of Experimental and Clinical Medicine , University of Florence , I-50134 Florence , Italy
| | - Maurizio Botta
- Dipartimento Biotecnologie, Chimica e Farmacia , Università degli Studi di Siena , Via A. De Gasperi 2 , I-53100 Siena , Italy.,Lead Discovery Siena S.r.l. , Castelnuovo Berardenga, I-53019 Siena , Italy.,Biotechnology College of Science and Technology , Temple University , BioLife Science Building, Suite 333, 1900 North 12th Street , Philadelphia , Pennsylvania 19122 , United States
| |
Collapse
|
34
|
Brennan R, Haap-Hoff A, Gu L, Gautier V, Long A, Schröder M. Investigating nucleo-cytoplasmic shuttling of the human DEAD-box helicase DDX3. Eur J Cell Biol 2018; 97:501-511. [PMID: 30131165 DOI: 10.1016/j.ejcb.2018.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 01/20/2023] Open
Abstract
The human DEAD-box helicase DDX3 is a multi-functional protein involved in the regulation of gene expression and additional non-conventional roles as signalling adaptor molecule that are independent of its enzymatic RNA remodeling activity. It is a nucleo-cytoplasmic shuttling protein and it has previously been suggested that dysregulation of its subcellular localization could contribute to tumourigenesis. Indeed, both tumour suppressor and oncogenic functions have been attributed to DDX3. In this study, we investigated the regulation of DDX3's nucleocytoplasmic shuttling. We confirmed that an N-terminal conserved Nuclear Export Signal (NES) is required for export of human DDX3 from the nucleus, and identified three regions within DDX3 that can independently facilitate its nuclear import. We also aimed to identify conditions that alter DDX3's subcellular localisation. Viral infection, cytokine treatment and DNA damage only induced minor changes in DDX3's subcellular distribution as determined by High Content Analysis. However, DDX3's nuclear localization increased in early mitotic cells (during prophase) concomitant with an increase in DDX3 expression levels. Our results are likely to have implications for the proposed use of (nuclear) DDX3 as a prognostic biomarker in cancer.
Collapse
Affiliation(s)
- Ruth Brennan
- Institute of Immunology, Biology Department, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Antje Haap-Hoff
- School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Lili Gu
- Institute of Immunology, Biology Department, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Virginie Gautier
- School of Medicine, Centre for Research in Infectious Diseases (CRID), University College Dublin, Belfield, Dublin 4, Ireland
| | - Aideen Long
- School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Martina Schröder
- Institute of Immunology, Biology Department, Maynooth University, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
35
|
Datta A, Brosh RM. New Insights Into DNA Helicases as Druggable Targets for Cancer Therapy. Front Mol Biosci 2018; 5:59. [PMID: 29998112 PMCID: PMC6028597 DOI: 10.3389/fmolb.2018.00059] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/06/2018] [Indexed: 12/28/2022] Open
Abstract
Small molecules that deter the functions of DNA damage response machinery are postulated to be useful for enhancing the DNA damaging effects of chemotherapy or ionizing radiation treatments to combat cancer by impairing the proliferative capacity of rapidly dividing cells that accumulate replicative lesions. Chemically induced or genetic synthetic lethality is a promising area in personalized medicine, but it remains to be optimized. A new target in cancer therapy is DNA unwinding enzymes known as helicases. Helicases play critical roles in all aspects of nucleic acid metabolism. We and others have investigated small molecule targeted inhibition of helicase function by compound screens using biochemical and cell-based approaches. Small molecule-induced trapping of DNA helicases may represent a generalized mechanism exemplified by certain topoisomerase and PARP inhibitors that exert poisonous consequences, especially in rapidly dividing cancer cells. Taking the lead from the broader field of DNA repair inhibitors and new information gleaned from structural and biochemical studies of DNA helicases, we predict that an emerging strategy to identify useful helicase-interacting compounds will be structure-based molecular docking interfaced with a computational approach. Potency, specificity, drug resistance, and bioavailability of helicase inhibitor drugs and targeting such compounds to subcellular compartments where the respective helicases operate must be addressed. Beyond cancer therapy, continued and new developments in this area may lead to the discovery of helicase-interacting compounds that chemically rescue clinically relevant helicase missense mutant proteins or activate the catalytic function of wild-type DNA helicases, which may have novel therapeutic application.
Collapse
Affiliation(s)
- Arindam Datta
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, NIH Biomedical Research Center, Baltimore, MD, United States
| | - Robert M Brosh
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, NIH Biomedical Research Center, Baltimore, MD, United States
| |
Collapse
|
36
|
Meier-Stephenson V, Mrozowich T, Pham M, Patel TR. DEAD-box helicases: the Yin and Yang roles in viral infections. Biotechnol Genet Eng Rev 2018; 34:3-32. [PMID: 29742983 DOI: 10.1080/02648725.2018.1467146] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Viruses hijack the host cell machinery and recruit host proteins to aid their replication. Several host proteins also play vital roles in inhibiting viral replication. Emerging class of host proteins central to both of these processes are the DEAD-box helicases: a highly conserved family of ATP-dependent RNA helicases, bearing a common D-E-A-D (Asp-Glu-Ala-Asp) motif. They play key roles in numerous cellular processes, including transcription, splicing, miRNA biogenesis and translation. Though their sequences are highly conserved, these helicases have quite diverse roles in the cell. Interestingly, often these helicases display contradictory actions in terms of the support and/or clearance of invading viruses. Increasing evidence highlights the importance of these enzymes, however, little is known about the structural basis of viral RNA recognition by the members of the DEAD-box family. This review summarizes the current knowledge in the field for selected DEAD-box helicases and highlights their diverse actions upon viral invasion of the host cell. We anticipate that through a better understanding of how these helicases are being utilized by viral RNAs and proteins to aid viral replication, it will be possible to address the urgent need to develop novel therapeutic approaches to combat viral infections.
Collapse
Affiliation(s)
- Vanessa Meier-Stephenson
- a Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute , University of Lethbridge , Lethbridge , Canada.,b Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine , University of Calgary , Calgary , Canada
| | - Tyler Mrozowich
- a Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute , University of Lethbridge , Lethbridge , Canada
| | - Mimi Pham
- a Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute , University of Lethbridge , Lethbridge , Canada
| | - Trushar R Patel
- a Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute , University of Lethbridge , Lethbridge , Canada.,b Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine , University of Calgary , Calgary , Canada.,c Faculty of Medicine & Dentistry, DiscoveryLab , University of Alberta , Edmonton , Canada
| |
Collapse
|
37
|
Cheng W, Chen G, Jia H, He X, Jing Z. DDX5 RNA Helicases: Emerging Roles in Viral Infection. Int J Mol Sci 2018; 19:ijms19041122. [PMID: 29642538 PMCID: PMC5979547 DOI: 10.3390/ijms19041122] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 02/07/2023] Open
Abstract
Asp-Glu-Ala-Asp (DEAD)-box polypeptide 5 (DDX5), also called p68, is a prototypical member of the large ATP-dependent RNA helicases family and is known to participate in all aspects of RNA metabolism ranging from transcription to translation, RNA decay, and miRNA processing. The roles of DDX5 in cell cycle regulation, tumorigenesis, apoptosis, cancer development, adipogenesis, Wnt-β-catenin signaling, and viral infection have been established. Several RNA viruses have been reported to hijack DDX5 to facilitate various steps of their replication cycles. Furthermore, DDX5 can be bounded by the viral proteins of some viruses with unknown functions. Interestingly, an antiviral function of DDX5 has been reported during hepatitis B virus and myxoma virus infection. Thus, the precise roles of this apparently multifaceted protein remain largely obscure. Here, we provide a rapid and critical overview of the structure and functions of DDX5 with a particular emphasis on its role during virus infection.
Collapse
Affiliation(s)
- Wenyu Cheng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Agriculture Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China.
| | - Guohua Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Agriculture Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China.
| | - Huaijie Jia
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Agriculture Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China.
| | - Xiaobing He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Agriculture Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China.
| | - Zhizhong Jing
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Agriculture Ministry, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China.
| |
Collapse
|
38
|
Cellular RNA Helicases Support Early and Late Events in Retroviral Replication. RETROVIRUS-CELL INTERACTIONS 2018. [PMCID: PMC7149973 DOI: 10.1016/b978-0-12-811185-7.00007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Retroviruses commandeer cell RNA helicases (RHs). Cell RHs are necessary for early and late events in retrovirus replication. The provirus is adopted by the cell-endogenous nuclear and cytoplasmic gene expression types of machinery. Whereas retroviruses engender the supportive activity of cell RHs, other RNA viruses provoke theantiviral role of this superfamily of conserved proteins. In this chapter, we contrast retrovirus reliance on host RNA helicases to support their replication cycle, with the virus-encoded helicaseactivity utilized by RNA viruses in cytoplasmic factories. Ironically, RHs are agonists to retroviruses and antagonists to other RNA viruses.
Collapse
|
39
|
Heerma van Voss MR, Vesuna F, Bol GM, Meeldijk J, Raman A, Offerhaus GJ, Buerger H, Patel AH, van der Wall E, van Diest PJ, Raman V. Nuclear DDX3 expression predicts poor outcome in colorectal and breast cancer. Onco Targets Ther 2017; 10:3501-3513. [PMID: 28761359 PMCID: PMC5522823 DOI: 10.2147/ott.s140639] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose DEAD box protein 3 (DDX3) is an RNA helicase with oncogenic properties that shuttles between the cytoplasm and nucleus. The majority of DDX3 is found in the cytoplasm, but a subset of tumors has distinct nuclear DDX3 localization of yet unknown biological significance. This study aimed to evaluate the significance of and mechanisms behind nuclear DDX3 expression in colorectal and breast cancer. Methods Expression of nuclear DDX3 and the nuclear exporter chromosome region maintenance 1 (CRM1) was evaluated by immunohistochemistry in 304 colorectal and 292 breast cancer patient samples. Correlations between the subcellular localization of DDX3 and CRM1 and the difference in overall survival between patients with and without nuclear DDX3 were studied. In addition, DDX3 mutants were created for in vitro evaluation of the mechanism behind nuclear retention of DDX3. Results DDX3 was present in the nucleus of 35% of colorectal and 48% of breast cancer patient samples and was particularly strong in the nucleolus. Nuclear DDX3 correlated with worse overall survival in both colorectal (hazard ratio [HR] 2.34, P<0.001) and breast cancer (HR 2.39, P=0.004) patients. Colorectal cancers with nuclear DDX3 expression more often had cytoplasmic expression of the nuclear exporter CRM1 (relative risk 1.67, P=0.04). In vitro analysis of DDX3 deletion mutants demonstrated that CRM1-mediated export was most dependent on the N-terminal nuclear export signal. Conclusion Overall, we conclude that nuclear DDX3 is partially CRM1-mediated and predicts worse survival in colorectal and breast cancer patients, putting it forward as a target for therapeutic intervention with DDX3 inhibitors under development in these cancer types.
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Farhad Vesuna
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guus M Bol
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jan Meeldijk
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ana Raman
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G Johan Offerhaus
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Arvind H Patel
- Centre for Virus Research, MRC-University of Glasgow, Glasgow, UK
| | - Elsken van der Wall
- Cancer Center, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Venu Raman
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
How to win the HIV-1 drug resistance hurdle race: running faster or jumping higher? Biochem J 2017; 474:1559-1577. [PMID: 28446620 DOI: 10.1042/bcj20160772] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/28/2017] [Accepted: 03/03/2017] [Indexed: 11/17/2022]
Abstract
Infections by the human immunodeficiency virus type 1 (HIV-1), the causative agent of the acquired immunodeficiency syndrome (AIDS), are still totaling an appalling 36.7 millions worldwide, with 1.1 million AIDS deaths/year and a similar number of yearly new infections. All this, in spite of the discovery of HIV-1 as the AIDS etiological agent more than 30 years ago and the introduction of an effective combinatorial antiretroviral therapy (cART), able to control disease progression, more than 20 years ago. Although very effective, current cART is plagued by the emergence of drug-resistant viral variants and most of the efforts in the development of novel direct-acting antiviral agents (DAAs) against HIV-1 have been devoted toward the fighting of resistance. In this review, rather than providing a detailed listing of all the drugs and the corresponding resistance mutations, we aim, through relevant examples, at presenting to the general reader the conceptual shift in the approaches that are being taken to overcome the viral resistance hurdle. From the classic 'running faster' strategy, based on the development of novel DAAs active against the mutant viruses selected by the previous drugs and/or presenting to the virus a high genetic barrier toward the development of resilience, to a 'jumping higher' approach, which looks at the cell, rather than the virus, as a source of valuable drug targets, in order to make the cellular environment non-permissive toward the replication of both wild-type and mutated viruses.
Collapse
|
41
|
Identification of novel cancer therapeutic targets using a designed and pooled shRNA library screen. Sci Rep 2017; 7:43023. [PMID: 28223711 PMCID: PMC5320502 DOI: 10.1038/srep43023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/17/2017] [Indexed: 01/08/2023] Open
Abstract
Targeted cancer therapeutics aim to exploit tumor-specific, genetic vulnerabilities specifically affecting neoplastic cells without similarly affecting normal cells. Here we performed sequencing-based screening of an shRNA library on a panel of cancer cells of different origins as well as normal cells. The shRNA library was designed to target a subset of genes previously identified using a whole genome screening approach. This focused shRNA library was infected into cells followed by analysis of enrichment and depletion of the shRNAs over the course of cell proliferation. We developed a bootstrap likelihood ratio test for the interpretation of the effects of multiple shRNAs over multiple cell line passages. Our analysis identified 44 genes whose depletion preferentially inhibited the growth of cancer cells. Among these genes ribosomal protein RPL35A, putative RNA helicase DDX24, and coatomer complex I (COPI) subunit ARCN1 most significantly inhibited growth of multiple cancer cell lines without affecting normal cell growth and survival. Further investigation revealed that the growth inhibition caused by DDX24 depletion is independent of p53 status underlining its value as a drug target. Overall, our study establishes a new approach for the analysis of proliferation-based shRNA selection strategies and identifies new targets for the development of cancer therapeutics.
Collapse
|
42
|
Tassini S, Sun L, Lanko K, Crespan E, Langron E, Falchi F, Kissova M, Armijos-Rivera JI, Delang L, Mirabelli C, Neyts J, Pieroni M, Cavalli A, Costantino G, Maga G, Vergani P, Leyssen P, Radi M. Discovery of Multitarget Agents Active as Broad-Spectrum Antivirals and Correctors of Cystic Fibrosis Transmembrane Conductance Regulator for Associated Pulmonary Diseases. J Med Chem 2017; 60:1400-1416. [DOI: 10.1021/acs.jmedchem.6b01521] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sabrina Tassini
- P4T Group, Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Liang Sun
- Laboratory
of Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Kristina Lanko
- Laboratory
of Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Emmanuele Crespan
- Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Emily Langron
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E
6BT London, U.K
| | - Federico Falchi
- CompuNet, Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
- Department of Pharmacy
and Biotechnology, University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Miroslava Kissova
- Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | | | - Leen Delang
- Laboratory
of Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Carmen Mirabelli
- Laboratory
of Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Johan Neyts
- Laboratory
of Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Marco Pieroni
- P4T Group, Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Andrea Cavalli
- CompuNet, Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy
- Department of Pharmacy
and Biotechnology, University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Gabriele Costantino
- P4T Group, Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Giovanni Maga
- Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Paola Vergani
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E
6BT London, U.K
| | - Pieter Leyssen
- Laboratory
of Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - Marco Radi
- P4T Group, Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| |
Collapse
|
43
|
Cai W, Xiong Chen Z, Rane G, Satendra Singh S, Choo Z, Wang C, Yuan Y, Zea Tan T, Arfuso F, Yap CT, Pongor LS, Yang H, Lee MB, Cher Goh B, Sethi G, Benoukraf T, Tergaonkar V, Prem Kumar A. Wanted DEAD/H or Alive: Helicases Winding Up in Cancers. J Natl Cancer Inst 2017; 109:2957323. [PMID: 28122908 DOI: 10.1093/jnci/djw278] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/08/2016] [Accepted: 10/20/2016] [Indexed: 12/23/2022] Open
Abstract
Cancer is one of the most studied areas of human biology over the past century. Despite having attracted much attention, hype, and investments, the search to find a cure for cancer remains an uphill battle. Recent discoveries that challenged the central dogma of molecular biology not only further increase the complexity but also demonstrate how various types of noncoding RNAs such as microRNA and long noncoding RNA, as well as their related processes such as RNA editing, are important in regulating gene expression. Parallel to this aspect, an increasing number of reports have focused on a family of proteins known as DEAD/H-box helicases involved in RNA metabolism, regulation of long and short noncoding RNAs, and novel roles as "editing helicases" and their association with cancers. This review summarizes recent findings on the roles of RNA helicases in various cancers, which are broadly classified into adult solid tumors, childhood solid tumors, leukemia, and cancer stem cells. The potential small molecule inhibitors of helicases and their therapeutic value are also discussed. In addition, analyzing next-generation sequencing data obtained from public portals and reviewing existing literature, we provide new insights on the potential of DEAD/H-box helicases to act as pharmacological drug targets in cancers.
Collapse
Affiliation(s)
- Wanpei Cai
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Zhi Xiong Chen
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Grishma Rane
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Shikha Satendra Singh
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Zhang'e Choo
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Chao Wang
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Yi Yuan
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Tuan Zea Tan
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Frank Arfuso
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Celestial T Yap
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Lorinc S Pongor
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Henry Yang
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Martin B Lee
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Boon Cher Goh
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Gautam Sethi
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Touati Benoukraf
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Vinay Tergaonkar
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Alan Prem Kumar
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| |
Collapse
|
44
|
Tuteja R. Emerging functions of helicases in regulation of stress survival in malaria parasite Plasmodium falciparum and their comparison with human host. Parasitol Int 2016; 65:645-664. [PMID: 27586396 DOI: 10.1016/j.parint.2016.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 06/28/2016] [Accepted: 08/28/2016] [Indexed: 02/04/2023]
Abstract
The cellular response to various stresses is a universal phenomenon and involves a common set of stress responses that are largely independent of the type of stress. The response to stress is complex and cells can activate multiple signaling pathways that act in concert to influence cell fate and results in a specific cellular outcome, including reduction in macromolecular synthesis by shared pathways, cell cycle arrest, DNA repair, senescence and/or apoptosis. Whether cells mount a protective response or die depends to a great degree on the nature and duration of the stress and the particular cell type. Helicases play essential roles in DNA replication, repair, recombination, transcription and translation, and also participate in RNA metabolic processes including pre-mRNA processing, ribosome biogenesis, RNA turnover, export, translation, surveillance, storage and decay. In order to survive in the human host, the malaria parasite Plasmodium falciparum has to handle variety of stresses, which it encounters during the erythrocytic stages of its life cycle. In recent past the role of helicases in imparting various stress responses has emerged. Therefore in the present review an attempt has been made to highlight the emerging importance of helicases in stress responses in malaria parasite and their comparison with human host is also presented. It is noteworthy that PfDHX33 and PfDDX60 are larger in size and different in sequence as compared to the HsDHX33 and HsDDX60. The study suggests that helicases are multifunctional and play major role in helping the cells to combat various stresses.
Collapse
Affiliation(s)
- Renu Tuteja
- Parasite Biology Group, International Centre for Genetic Engineering and Biotechnology, P. O. Box 10504, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
45
|
Xie M, Vesuna F, Botlagunta M, Bol GM, Irving A, Bergman Y, Hosmane RS, Kato Y, Winnard PT, Raman V. NZ51, a ring-expanded nucleoside analog, inhibits motility and viability of breast cancer cells by targeting the RNA helicase DDX3. Oncotarget 2016; 6:29901-13. [PMID: 26337079 PMCID: PMC4745771 DOI: 10.18632/oncotarget.4898] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/29/2015] [Indexed: 12/21/2022] Open
Abstract
DDX3X (DDX3), a human RNA helicase, is over expressed in multiple breast cancer cell lines and its expression levels are directly correlated to cellular aggressiveness. NZ51, a ring-expanded nucleoside analogue (REN) has been reported to inhibit the ATP dependent helicase activity of DDX3. Molecular modeling of NZ51 binding to DDX3 indicated that the 5:7-fused imidazodiazepine ring of NZ51 was incorporated into the ATP binding pocket of DDX3. In this study, we investigated the anticancer properties of NZ51 in MCF-7 and MDA-MB-231 breast cancer cell lines. NZ51 treatment decreased cellular motility and cell viability of MCF-7 and MDA-MB-231 cells with IC50 values in the low micromolar range. Biological knockdown of DDX3 in MCF-7 and MDA-MB-231 cells resulted in decreased proliferation rates and reduced clonogenicity. In addition, NZ51 was effective in killing breast cancer cells under hypoxic conditions with the same potency as observed during normoxia. Mechanistic studies indicated that NZ51 did not cause DDX3 degradation, but greatly diminished its functionality. Moreover, in vivo experiments demonstrated that DDX3 knockdown by shRNA resulted in reduced tumor volume and metastasis without altering tumor vascular volume or permeability-surface area. In initial in vivo experiments, NZ51 treatment did not significantly reduce tumor volume. Further studies are needed to optimize drug formulation, dose and delivery. Continuing work will determine the in vitro-in vivo correlation of NZ51 activity and its utility in a clinical setting.
Collapse
Affiliation(s)
- Min Xie
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Farhad Vesuna
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mahendran Botlagunta
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guus Martinus Bol
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, University Medical Center Utrecht Cancer Center, GA, Utrecht, The Netherlands
| | - Ashley Irving
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yehudit Bergman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ramachandra S Hosmane
- Department of Chemistry & Biochemistry, University of Maryland, Baltimore County, MD, USA
| | - Yoshinori Kato
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul T Winnard
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Venu Raman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, University Medical Center Utrecht Cancer Center, GA, Utrecht, The Netherlands.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
46
|
Fröhlich A, Rojas-Araya B, Pereira-Montecinos C, Dellarossa A, Toro-Ascuy D, Prades-Pérez Y, García-de-Gracia F, Garcés-Alday A, Rubilar PS, Valiente-Echeverría F, Ohlmann T, Soto-Rifo R. DEAD-box RNA helicase DDX3 connects CRM1-dependent nuclear export and translation of the HIV-1 unspliced mRNA through its N-terminal domain. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1859:719-30. [PMID: 27012366 DOI: 10.1016/j.bbagrm.2016.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/14/2016] [Accepted: 03/17/2016] [Indexed: 12/20/2022]
Abstract
DEAD-box RNA helicase DDX3 is a host factor essential for HIV-1 replication and thus, a potential target for novel therapies aimed to overcome viral resistance. Previous studies have shown that DDX3 promotes nuclear export and translation of the HIV-1 unspliced mRNA. Although the function of DDX3 during both processes requires its catalytic activity, it is unknown whether other domains surrounding the helicase core are involved. Here, we show the involvement of the N- and C-terminal domains of DDX3 in the regulation of HIV-1 unspliced mRNA translation. Our results suggest that the intrinsically disordered N-terminal domain of DDX3 regulates its functions in translation by acting prior to the recruitment of the 43S pre-initiation complex onto the viral 5'-UTR. Interestingly, this regulation was conserved in HIV-2 and was dependent on the CRM1-dependent nuclear export pathway suggesting a role of the RNA helicase in interconnecting nuclear export with ribosome recruitment of the viral unspliced mRNA. This specific function of DDX3 during HIV gene expression could be exploited as an alternative target for pharmaceutical intervention.
Collapse
Affiliation(s)
- Alvaro Fröhlich
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Bárbara Rojas-Araya
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Camila Pereira-Montecinos
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Alessandra Dellarossa
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Daniela Toro-Ascuy
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Yara Prades-Pérez
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Francisco García-de-Gracia
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Andrea Garcés-Alday
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Paulina S Rubilar
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France
| | - Fernando Valiente-Echeverría
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile
| | - Théophile Ohlmann
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France
| | - Ricardo Soto-Rifo
- Molecular and Cellular Virology Laboratory, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 834100, Santiago, Chile.
| |
Collapse
|
47
|
Human DDX3 protein is a valuable target to develop broad spectrum antiviral agents. Proc Natl Acad Sci U S A 2016; 113:5388-93. [PMID: 27118832 DOI: 10.1073/pnas.1522987113] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Targeting a host factor essential for the replication of different viruses but not for the cells offers a higher genetic barrier to the development of resistance, may simplify therapy regimens for coinfections, and facilitates management of emerging viral diseases. DEAD-box polypeptide 3 (DDX3) is a human host factor required for the replication of several DNA and RNA viruses, including some of the most challenging human pathogens currently circulating, such as HIV-1, Hepatitis C virus, Dengue virus, and West Nile virus. Herein, we showed for the first time, to our knowledge, that the inhibition of DDX3 by a small molecule could be successfully exploited for the development of a broad spectrum antiviral agent. In addition to the multiple antiviral activities, hit compound 16d retained full activity against drug-resistant HIV-1 strains in the absence of cellular toxicity. Pharmacokinetics and toxicity studies in rats confirmed a good safety profile and bioavailability of 16d. Thus, DDX3 is here validated as a valuable therapeutic target.
Collapse
|
48
|
Suthar MK, Purva M, Maherchandani S, Kashyap SK. Identification and in silico analysis of cattle DExH/D box RNA helicases. SPRINGERPLUS 2016; 5:25. [PMID: 26783509 PMCID: PMC4705078 DOI: 10.1186/s40064-015-1640-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/20/2015] [Indexed: 12/16/2022]
Abstract
The helicases are motor proteins participating
in a range of nucleic acid metabolisms. RNA helicase families are characterized by the presence of conserved motifs. This article reports a comprehensive in silico analysis of Bos taurus DExH/D helicase members. Bovine helicases were identified using the helicase domain sequences including 38 DDX (DEAD box) and 16 DHX (DEAH box) members. Signature motifs were used for the validation of these proteins. Putative sub cellular localization and phylogenetic relationship for these RNA helicases were established. Comparative analysis of these proteins with human DDX and DHX members was carried out. These bovine helicase have been assigned putative physiological functions. Present study of cattle DExH/D helicase will provides an invaluable source for the detailed biochemical and physiological research on these members.
Collapse
Affiliation(s)
- Manish Kumar Suthar
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary & Animal Sciences, Bikaner, Rajasthan 334001 India
| | - Mukul Purva
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary & Animal Sciences, Bikaner, Rajasthan 334001 India
| | - Sunil Maherchandani
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary & Animal Sciences, Bikaner, Rajasthan 334001 India
| | - Sudhir Kumar Kashyap
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary & Animal Sciences, Bikaner, Rajasthan 334001 India
| |
Collapse
|
49
|
Fazi R, Tintori C, Brai A, Botta L, Selvaraj M, Garbelli A, Maga G, Botta M. Homology Model-Based Virtual Screening for the Identification of Human Helicase DDX3 Inhibitors. J Chem Inf Model 2015; 55:2443-54. [PMID: 26544088 DOI: 10.1021/acs.jcim.5b00419] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Targeting cellular cofactors instead of viral enzymes represents a new strategy to combat infectious diseases, which should help to overcome the problem of viral resistance. Recently, it has been revealed that the cellular ATPase/RNA helicase X-linked DEAD-box polypeptide 3 (DDX3) is an essential host factor for the replication of several viruses such as HIV, HCV, JEV, Dengue, and West Nile. Accordingly, a drug targeting DDX3 could theoretically inhibit all viruses that are dependent on this host factor. Herein, for the first time, a model of hDDX3 in its closed conformation, which binds the viral RNA was developed by using the homology module of Prime through the Maestro interface of Schrodinger. Next, a structure-based virtual screening protocol was applied to identify DDX3 small molecule inhibitors targeting the RNA binding pocket. As a result, an impressive hit rate of 40% was obtained with the identification of 10 active compounds out of the 25 tested small molecules. The best poses of the active ligands highlighted the crucial residues to be targeted for the inhibition of the helicase activity of DDX3. The obtained results confirm the reliability of the constructed DDX3/RNA model and the proposed computational strategy for investigating novel DDX3 inhibitors.
Collapse
Affiliation(s)
- Roberta Fazi
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena , Via A. De Gasperi 2, I-53100 Siena, Italy
| | - Cristina Tintori
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena , Via A. De Gasperi 2, I-53100 Siena, Italy
| | - Annalaura Brai
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena , Via A. De Gasperi 2, I-53100 Siena, Italy
| | - Lorenzo Botta
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena , Via A. De Gasperi 2, I-53100 Siena, Italy
| | - Manikandan Selvaraj
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena , Via A. De Gasperi 2, I-53100 Siena, Italy
| | - Anna Garbelli
- Istituto di Genetica Molecolare, IGM-CNR , Via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Giovanni Maga
- Istituto di Genetica Molecolare, IGM-CNR , Via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Maurizio Botta
- Dipartimento Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena , Via A. De Gasperi 2, I-53100 Siena, Italy.,Biotechnology College of Science and Technology, Temple University , Biolife Science Building, Suite 333, 1900 N 12th Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
50
|
Bol GM, Xie M, Raman V. DDX3, a potential target for cancer treatment. Mol Cancer 2015; 14:188. [PMID: 26541825 PMCID: PMC4636063 DOI: 10.1186/s12943-015-0461-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/22/2015] [Indexed: 12/27/2022] Open
Abstract
RNA helicases are a large family of proteins with a distinct motif, referred to as the DEAD/H (Asp-Glu-Ala-Asp/His). The exact functions of all the human DEAD/H box proteins are unknown. However, it has been consistently demonstrated that these proteins are associated with several aspects of energy-dependent RNA metabolism, including translation, ribosome biogenesis, and pre-mRNA splicing. In addition, DEAD/H box proteins participate in nuclear-cytoplasmic transport and organellar gene expression. A member of this RNA helicase family, DDX3, has been identified in a variety of cellular biogenesis processes, including cell-cycle regulation, cellular differentiation, cell survival, and apoptosis. In cancer, DDX3 expression has been evaluated in patient samples of breast, lung, colon, oral, and liver cancer. Both tumor suppressor and oncogenic functions have been attributed to DDX3 and are discussed in this review. In general, there is concordance with in vitro evidence to support the hypothesis that DDX3 is associated with an aggressive phenotype in human malignancies. Interestingly, very few cancer types harbor mutations in DDX3, which result in altered protein function rather than a loss of function. Efficacy of drugs to curtail cancer growth is hindered by adaptive responses that promote drug resistance, eventually leading to treatment failure. One way to circumvent development of resistant disease is to develop novel drugs that target over-expressed proteins involved in this adaptive response. Moreover, if the target gene is developmentally regulated, there is less of a possibility to abruptly accumulate mutations leading to drug resistance. In this regard, DDX3 could be a druggable target for cancer treatment. We present an overview of DDX3 biology and the currently available DDX3 inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Guus Martinus Bol
- Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA, Utrecht, The Netherlands.,Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Ave, Traylor 340, Baltimore, MD, 21205, USA
| | - Min Xie
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Ave, Traylor 340, Baltimore, MD, 21205, USA
| | - Venu Raman
- Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA, Utrecht, The Netherlands. .,Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 720 Rutland Ave, Traylor 340, Baltimore, MD, 21205, USA. .,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|