1
|
Stratigi K, Siametis A, Garinis GA. Looping forward: exploring R-loop processing and therapeutic potential. FEBS Lett 2025; 599:244-266. [PMID: 38844597 PMCID: PMC11771710 DOI: 10.1002/1873-3468.14947] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 01/28/2025]
Abstract
Recently, there has been increasing interest in the complex relationship between transcription and genome stability, with specific attention directed toward the physiological significance of molecular structures known as R-loops. These structures arise when an RNA strand invades into the DNA duplex, and their formation is involved in a wide range of regulatory functions affecting gene expression, DNA repair processes or cell homeostasis. The persistent presence of R-loops, if not effectively removed, contributes to genome instability, underscoring the significance of the factors responsible for their resolution and modification. In this review, we provide a comprehensive overview of how R-loop processing can drive either a beneficial or a harmful outcome. Additionally, we explore the potential for manipulating such structures to devise rationalized therapeutic strategies targeting the aberrant accumulation of R-loops.
Collapse
Affiliation(s)
- Kalliopi Stratigi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
| | - Athanasios Siametis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
- Department of BiologyUniversity of CreteHeraklionCreteGreece
| | - George A. Garinis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
- Department of BiologyUniversity of CreteHeraklionCreteGreece
| |
Collapse
|
2
|
Obermann R, Yemane B, Jarvis C, Franco FM, Kyriukha Y, Nolan W, Gohara B, Krezel AM, Wildman SA, Janetka JW. Small Molecule Antagonists of the DNA Repair ERCC1/XPA Protein-Protein Interaction. ChemMedChem 2024; 19:e202300648. [PMID: 38300970 PMCID: PMC11031295 DOI: 10.1002/cmdc.202300648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/03/2024]
Abstract
The DNA excision repair protein ERCC1 and the DNA damage sensor protein, XPA are highly overexpressed in patient samples of cisplatin-resistant solid tumors including lung, bladder, ovarian, and testicular cancer. The repair of cisplatin-DNA crosslinks is dependent upon nucleotide excision repair (NER) that is modulated by protein-protein binding interactions of ERCC1, the endonuclease, XPF, and XPA. Thus, inhibition of their function is a potential therapeutic strategy for the selective sensitization of tumors to DNA-damaging platinum-based cancer therapy. Here, we report on new small-molecule antagonists of the ERCC1/XPA protein-protein interaction (PPI) discovered using a high-throughput competitive fluorescence polarization binding assay. We discovered a unique structural class of thiopyridine-3-carbonitrile PPI antagonists that block a truncated XPA polypeptide from binding to ERCC1. Preliminary hit-to-lead studies from compound 1 reveal structure-activity relationships (SAR) and identify lead compound 27 o with an EC50 of 4.7 μM. Furthermore, chemical shift perturbation mapping by NMR confirms that 1 binds within the same site as the truncated XPA67-80 peptide. These novel ERCC1 antagonists are useful chemical biology tools for investigating DNA damage repair pathways and provide a good starting point for medicinal chemistry optimization as therapeutics for sensitizing tumors to DNA damaging agents and overcoming resistance to platinum-based chemotherapy.
Collapse
Affiliation(s)
| | | | - Cassie Jarvis
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Francisco M. Franco
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Yevhenii Kyriukha
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - William Nolan
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Beth Gohara
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Andrzej M. Krezel
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Scott A. Wildman
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - James W. Janetka
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| |
Collapse
|
3
|
eIF3a regulation of mTOR signaling and translational control via HuR in cellular response to DNA damage. Oncogene 2022; 41:2431-2443. [PMID: 35279705 PMCID: PMC9035104 DOI: 10.1038/s41388-022-02262-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 01/29/2023]
Abstract
eIF3a (eukaryotic translation initiation factor 3a), a subunit of the eIF3 complex, has been suggested to play a regulatory role in protein synthesis and in cellular response to DNA-damaging treatments. S6K1 is an effector and a mediator of mTOR complex 1 (mTORC1) in regulating protein synthesis and integrating diverse signals into control of cell growth and response to stress. Here, we show that eIF3a regulates S6K1 activity by inhibiting mTORC1 kinase via regulating Raptor synthesis. The regulation of Raptor synthesis is via eIF3a interaction with HuR (human antigen R) and binding of the eIF3a-HuR complex to the 5'-UTR of Raptor mRNA. Furthermore, mTORC1 may mediate eIF3a function in cellular response to cisplatin by regulating synthesis of NER proteins and NER activity. Taken together, we conclude that the mTOR signaling pathway may also be regulated by translational control and mediate eIF3a regulation of cancer cell response to cisplatin by regulating NER protein synthesis.
Collapse
|
4
|
VanderVere-Carozza PS, Gavande NS, Jalal SI, Pollok KE, Ekinci E, Heyza J, Patrick SM, Masters A, Turchi JJ, Pawelczak KS. In Vivo Targeting Replication Protein A for Cancer Therapy. Front Oncol 2022; 12:826655. [PMID: 35251993 PMCID: PMC8895377 DOI: 10.3389/fonc.2022.826655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Replication protein A (RPA) plays essential roles in DNA replication, repair, recombination, and the DNA damage response (DDR). Retrospective analysis of lung cancer patient data demonstrates high RPA expression as a negative prognostic biomarker for overall survival in smoking-related lung cancers. Similarly, relative expression of RPA is a predictive marker for response to chemotherapy. These observations are consistent with the increase in RPA expression serving as an adaptive mechanism that allows tolerance of the genotoxic stress resulting from carcinogen exposure. We have developed second-generation RPA inhibitors (RPAis) that block the RPA-DNA interaction and optimized formulation for in vivo analyses. Data demonstrate that unlike first-generation RPAis, second-generation molecules show increased cellular permeability and induce cell death via apoptosis. Second-generation RPAis elicit single-agent in vitro anticancer activity across a broad spectrum of cancers, and the cellular response suggests existence of a threshold before chemical RPA exhaustion induces cell death. Chemical RPA inhibition potentiates the anticancer activity of a series of DDR inhibitors and traditional DNA-damaging cancer therapeutics. Consistent with chemical RPA exhaustion, we demonstrate that the effects of RPAi on replication fork dynamics are similar to other known DDR inhibitors. An optimized formulation of RPAi NERx 329 was developed that resulted in single-agent anticancer activity in two non-small cell lung cancer models. These data demonstrate a unique mechanism of action of RPAis eliciting a state of chemical RPA exhaustion and suggest they will provide an effective therapeutic option for difficult-to-treat lung cancers.
Collapse
Affiliation(s)
| | - Navnath S. Gavande
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States,Department of Pharmaceutical Sciences, Wayne State University College of Pharmacy and Health Sciences, Detroit, MI, United States
| | - Shadia I. Jalal
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Karen E. Pollok
- Herman B. Wells Center for Pediatric Research, Departments of Pediatrics, Pharmacology and Toxicology, Medical and Molecular Genetics Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Elmira Ekinci
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
| | - Joshua Heyza
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
| | - Steve M. Patrick
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
| | - Andi Masters
- Indiana University Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John J. Turchi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States,NERx BioSciences, Indianapolis, IN, United States,*Correspondence: John J. Turchi, ; Katherine S. Pawelczak,
| | - Katherine S. Pawelczak
- NERx BioSciences, Indianapolis, IN, United States,*Correspondence: John J. Turchi, ; Katherine S. Pawelczak,
| |
Collapse
|
5
|
Targeting DNA Damage Response and Repair to Enhance Therapeutic Index in Cisplatin-Based Cancer Treatment. Int J Mol Sci 2021; 22:ijms22158199. [PMID: 34360968 PMCID: PMC8347825 DOI: 10.3390/ijms22158199] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Platinum-based chemotherapies, such as cisplatin, play a large role in cancer treatment. The development of resistance and treatment toxicity creates substantial barriers to disease control, yet. To enhance the therapeutic index of cisplatin-based chemotherapy, it is imperative to circumvent resistance and toxicity while optimizing tumor sensitization. One of the primary mechanisms by which cancer cells develop resistance to cisplatin is through upregulation of DNA repair pathways. In this review, we discuss the DNA damage response in the context of cisplatin-induced DNA damage. We describe the proteins involved in the pathways and their roles in resistance development. Common biomarkers for cisplatin resistance and their utilization to improve patient risk stratification and treatment personalization are addressed. Finally, we discuss some of the current treatments and future strategies to circumvent the development of cisplatin resistance.
Collapse
|
6
|
Role of Nucleotide Excision Repair in Cisplatin Resistance. Int J Mol Sci 2020; 21:ijms21239248. [PMID: 33291532 PMCID: PMC7730652 DOI: 10.3390/ijms21239248] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is a chemotherapeutic drug used for the treatment of a number of cancers. The efficacy of cisplatin relies on its binding to DNA and the induction of cytotoxic DNA damage to kill cancer cells. Cisplatin-based therapy is best known for curing testicular cancer; however, treatment of other solid tumors with cisplatin has not been as successful. Pre-clinical and clinical studies have revealed nucleotide excision repair (NER) as a major resistance mechanism against cisplatin in tumor cells. NER is a versatile DNA repair system targeting a wide range of helix-distorting DNA damage. The NER pathway consists of multiple steps, including damage recognition, pre-incision complex assembly, dual incision, and repair synthesis. NER proteins can recognize cisplatin-induced DNA damage and remove the damage from the genome, thereby neutralizing the cytotoxicity of cisplatin and causing drug resistance. Here, we review the molecular mechanism by which NER repairs cisplatin damage, focusing on the recent development of genome-wide cisplatin damage mapping methods. We also discuss how the expression and somatic mutations of key NER genes affect the response of cancer cells to cisplatin. Finally, small molecules targeting NER factors provide important tools to manipulate NER capacity in cancer cells. The status of research on these inhibitors and their implications in cancer treatment will be discussed.
Collapse
|
7
|
Interplay between Cellular Metabolism and the DNA Damage Response in Cancer. Cancers (Basel) 2020; 12:cancers12082051. [PMID: 32722390 PMCID: PMC7463900 DOI: 10.3390/cancers12082051] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
Metabolism is a fundamental cellular process that can become harmful for cells by leading to DNA damage, for instance by an increase in oxidative stress or through the generation of toxic byproducts. To deal with such insults, cells have evolved sophisticated DNA damage response (DDR) pathways that allow for the maintenance of genome integrity. Recent years have seen remarkable progress in our understanding of the diverse DDR mechanisms, and, through such work, it has emerged that cellular metabolic regulation not only generates DNA damage but also impacts on DNA repair. Cancer cells show an alteration of the DDR coupled with modifications in cellular metabolism, further emphasizing links between these two fundamental processes. Taken together, these compelling findings indicate that metabolic enzymes and metabolites represent a key group of factors within the DDR. Here, we will compile the current knowledge on the dynamic interplay between metabolic factors and the DDR, with a specific focus on cancer. We will also discuss how recently developed high-throughput technologies allow for the identification of novel crosstalk between the DDR and metabolism, which is of crucial importance to better design efficient cancer treatments.
Collapse
|
8
|
Leong SX, Koh LK, Koh CSL, Phan-Quang GC, Lee HK, Ling XY. In Situ Differentiation of Multiplex Noncovalent Interactions Using SERS and Chemometrics. ACS APPLIED MATERIALS & INTERFACES 2020; 12:33421-33427. [PMID: 32578974 DOI: 10.1021/acsami.0c08053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Probing changes of noncovalent interactions is crucial to study the binding efficiencies and strengths of (bio)molecular complexes. While surface-enhanced Raman scattering (SERS) offers unique molecular fingerprints to examine such interactions in situ, current platforms are only able to recognize hydrogen bonds because of their reliance on manual spectral identification. Here, we differentiate multiple intermolecular interactions between two interacting species by synergizing plasmonic liquid marble-based SERS platforms, chemometrics, and density functional theory. We demonstrate that characteristic 3-mercaptobenzoic acid (probe) Raman signals have distinct peak shifts upon hydrogen bonding and ionic interactions with tert-butylamine, a model interacting species. Notably, we further quantify the contributions from each noncovalent interaction coexisting in different proportions. As a proof-of-concept, we detect and categorize biologically important nucleotide bases based on molecule-specific interactions. This will potentially be useful to study how subtle changes in biomolecular interactions affect their structural and binding properties.
Collapse
Affiliation(s)
- Shi Xuan Leong
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Li Keng Koh
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Charlynn Sher Lin Koh
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Gia Chuong Phan-Quang
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Hiang Kwee Lee
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Xing Yi Ling
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| |
Collapse
|
9
|
XPA: DNA Repair Protein of Significant Clinical Importance. Int J Mol Sci 2020; 21:ijms21062182. [PMID: 32235701 PMCID: PMC7139726 DOI: 10.3390/ijms21062182] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023] Open
Abstract
The nucleotide excision repair (NER) pathway is activated in response to a broad spectrum of DNA lesions, including bulky lesions induced by platinum-based chemotherapeutic agents. Expression levels of NER factors and resistance to chemotherapy has been examined with some suggestion that NER plays a role in tumour resistance; however, there is a great degree of variability in these studies. Nevertheless, recent clinical studies have suggested Xeroderma Pigmentosum group A (XPA) protein, a key regulator of the NER pathway that is essential for the repair of DNA damage induced by platinum-based chemotherapeutics, as a potential prognostic and predictive biomarker for response to treatment. XPA functions in damage verification step in NER, as well as a molecular scaffold to assemble other NER core factors around the DNA damage site, mediated by protein–protein interactions. In this review, we focus on the interacting partners and mechanisms of regulation of the XPA protein. We summarize clinical oncology data related to this DNA repair factor, particularly its relationship with treatment outcome, and examine the potential of XPA as a target for small molecule inhibitors.
Collapse
|
10
|
|
11
|
Identification of small molecule inhibitors of ERCC1-XPF that inhibit DNA repair and potentiate cisplatin efficacy in cancer cells. Oncotarget 2018; 7:75104-75117. [PMID: 27650543 PMCID: PMC5342726 DOI: 10.18632/oncotarget.12072] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/02/2016] [Indexed: 12/11/2022] Open
Abstract
ERCC1-XPF heterodimer is a 5′-3′ structure-specific endonuclease which is essential in multiple DNA repair pathways in mammalian cells. ERCC1-XPF (ERCC1-ERCC4) repairs cisplatin-DNA intrastrand adducts and interstrand crosslinks and its specific inhibition has been shown to enhance cisplatin cytotoxicity in cancer cells. In this study, we describe a high throughput screen (HTS) used to identify small molecules that inhibit the endonuclease activity of ERCC1-XPF. Primary screens identified two compounds that inhibit ERCC1-XPF activity in the nanomolar range. These compounds were validated in secondary screens against two other non-related endonucleases to ensure specificity. Results from these screens were validated using an in vitro gel-based nuclease assay. Electrophoretic mobility shift assays (EMSAs) further show that these compounds do not inhibit the binding of purified ERCC1-XPF to DNA. Next, in lung cancer cells these compounds potentiated cisplatin cytotoxicity and inhibited DNA repair. Structure activity relationship (SAR) studies identified related compounds for one of the original Hits, which also potentiated cisplatin cytotoxicity in cancer cells. Excitingly, dosing with NSC16168 compound potentiated cisplatin antitumor activity in a lung cancer xenograft model. Further development of ERCC1-XPF DNA repair inhibitors is expected to sensitize cancer cells to DNA damage-based chemotherapy.
Collapse
|
12
|
Diouf B, Lin W, Goktug A, Grace CRR, Waddell MB, Bao J, Shao Y, Heath RJ, Zheng JJ, Shelat AA, Relling MV, Chen T, Evans WE. Alteration of RNA Splicing by Small-Molecule Inhibitors of the Interaction between NHP2L1 and U4. SLAS DISCOVERY 2017; 23:164-173. [PMID: 28985478 DOI: 10.1177/2472555217735035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Splicing is an important eukaryotic mechanism for expanding the transcriptome and proteome, influencing a number of biological processes. Understanding its regulation and identifying small molecules that modulate this process remain a challenge. We developed an assay based on time-resolved fluorescence resonance energy transfer (TR-FRET) to detect the interaction between the protein NHP2L1 and U4 RNA, which are two key components of the spliceosome. We used this assay to identify small molecules that interfere with this interaction in a high-throughput screening (HTS) campaign. Topotecan and other camptothecin derivatives were among the top hits. We confirmed that topotecan disrupts the interaction between NHP2L1 and U4 by binding to U4 and inhibits RNA splicing. Our data reveal new functions of known drugs that could facilitate the development of therapeutic strategies to modify splicing and alter gene function.
Collapse
Affiliation(s)
- Barthelemy Diouf
- 1 Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.,2 Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wenwei Lin
- 3 Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Asli Goktug
- 3 Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christy R R Grace
- 4 Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael Brett Waddell
- 5 Molecular Interaction Analysis Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ju Bao
- 1 Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.,2 Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Youming Shao
- 6 Protein Production Facility, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard J Heath
- 6 Protein Production Facility, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jie J Zheng
- 7 Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Anang A Shelat
- 3 Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mary V Relling
- 1 Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.,2 Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taosheng Chen
- 3 Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - William E Evans
- 1 Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.,2 Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
13
|
Gavande NS, VanderVere-Carozza P, Mishra AK, Vernon TL, Pawelczak KS, Turchi JJ. Design and Structure-Guided Development of Novel Inhibitors of the Xeroderma Pigmentosum Group A (XPA) Protein-DNA Interaction. J Med Chem 2017; 60:8055-8070. [PMID: 28933851 DOI: 10.1021/acs.jmedchem.7b00780] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
XPA is a unique and essential protein required for the nucleotide excision DNA repair pathway and represents a therapeutic target in oncology. Herein, we are the first to develop novel inhibitors of the XPA-DNA interaction through structure-guided drug design efforts. Ester derivatives of the compounds 1 (X80), 22, and 24 displayed excellent inhibitory activity (IC50 of 0.82 ± 0.18 μM and 1.3 ± 0.22 μM, respectively) but poor solubility. We have synthesized novel amide derivatives that retain potency and have much improved solubility. Furthermore, compound 1 analogs exhibited good specificity for XPA over RPA (replication protein A), another DNA-binding protein that participates in the nucleotide excision repair (NER) pathway. Importantly, there were no significant interactions observed by the X80 class of compounds directly with DNA. Molecular docking studies revealed a mechanistic model for the interaction, and these studies could serve as the basis for continued analysis of structure-activity relationships and drug development efforts of this novel target.
Collapse
Affiliation(s)
- Navnath S Gavande
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Pamela VanderVere-Carozza
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Akaash K Mishra
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Tyler L Vernon
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States
| | - Katherine S Pawelczak
- NERx Biosciences , 212 W 10th Street, Suite A480, Indianapolis, Indiana 46202, United States
| | - John J Turchi
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine , Indianapolis, Indiana 46202, United States.,NERx Biosciences , 212 W 10th Street, Suite A480, Indianapolis, Indiana 46202, United States
| |
Collapse
|
14
|
Nickoloff JA, Jones D, Lee SH, Williamson EA, Hromas R. Drugging the Cancers Addicted to DNA Repair. J Natl Cancer Inst 2017; 109:3832892. [PMID: 28521333 PMCID: PMC5436301 DOI: 10.1093/jnci/djx059] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/10/2017] [Indexed: 12/29/2022] Open
Abstract
Defects in DNA repair can result in oncogenic genomic instability. Cancers occurring from DNA repair defects were once thought to be limited to rare inherited mutations (such as BRCA1 or 2). It now appears that a clinically significant fraction of cancers have acquired DNA repair defects. DNA repair pathways operate in related networks, and cancers arising from loss of one DNA repair component typically become addicted to other repair pathways to survive and proliferate. Drug inhibition of the rescue repair pathway prevents the repair-deficient cancer cell from replicating, causing apoptosis (termed synthetic lethality). However, the selective pressure of inhibiting the rescue repair pathway can generate further mutations that confer resistance to the synthetic lethal drugs. Many such drugs currently in clinical use inhibit PARP1, a repair component to which cancers arising from inherited BRCA1 or 2 mutations become addicted. It is now clear that drugs inducing synthetic lethality may also be therapeutic in cancers with acquired DNA repair defects, which would markedly broaden their applicability beyond treatment of cancers with inherited DNA repair defects. Here we review how each DNA repair pathway can be attacked therapeutically and evaluate DNA repair components as potential drug targets to induce synthetic lethality. Clinical use of drugs targeting DNA repair will markedly increase when functional and genetic loss of repair components are consistently identified. In addition, future therapies will exploit artificial synthetic lethality, where complementary DNA repair pathways are targeted simultaneously in cancers without DNA repair defects.
Collapse
Affiliation(s)
- Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Dennie Jones
- Department of Medicine and the Cancer Center, University of Florida Health, Gainesville, FL, USA
| | - Suk-Hee Lee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth A Williamson
- Department of Medicine and the Cancer Center, University of Florida Health, Gainesville, FL, USA
| | - Robert Hromas
- Department of Medicine and the Cancer Center, University of Florida Health, Gainesville, FL, USA
| |
Collapse
|
15
|
Sugitani N, Sivley RM, Perry KE, Capra JA, Chazin WJ. XPA: A key scaffold for human nucleotide excision repair. DNA Repair (Amst) 2016; 44:123-135. [PMID: 27247238 DOI: 10.1016/j.dnarep.2016.05.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nucleotide excision repair (NER) is essential for removing many types of DNA lesions from the genome, yet the mechanisms of NER in humans remain poorly understood. This review summarizes our current understanding of the structure, biochemistry, interaction partners, mechanisms, and disease-associated mutations of one of the critical NER proteins, XPA.
Collapse
Affiliation(s)
- Norie Sugitani
- Departments of Biochemistry, Biological Sciences, Biomedical Informatics, Chemistry, and Computer Science, and Vanderbilt Genetics Institute and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232-7917, United States
| | - Robert M Sivley
- Departments of Biochemistry, Biological Sciences, Biomedical Informatics, Chemistry, and Computer Science, and Vanderbilt Genetics Institute and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232-7917, United States
| | - Kelly E Perry
- Departments of Biochemistry, Biological Sciences, Biomedical Informatics, Chemistry, and Computer Science, and Vanderbilt Genetics Institute and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232-7917, United States
| | - John A Capra
- Departments of Biochemistry, Biological Sciences, Biomedical Informatics, Chemistry, and Computer Science, and Vanderbilt Genetics Institute and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232-7917, United States
| | - Walter J Chazin
- Departments of Biochemistry, Biological Sciences, Biomedical Informatics, Chemistry, and Computer Science, and Vanderbilt Genetics Institute and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232-7917, United States.
| |
Collapse
|
16
|
Gavande NS, VanderVere-Carozza PS, Hinshaw HD, Jalal SI, Sears CR, Pawelczak KS, Turchi JJ. DNA repair targeted therapy: The past or future of cancer treatment? Pharmacol Ther 2016; 160:65-83. [PMID: 26896565 DOI: 10.1016/j.pharmthera.2016.02.003] [Citation(s) in RCA: 272] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The repair of DNA damage is a complex process that relies on particular pathways to remedy specific types of damage to DNA. The range of insults to DNA includes small, modest changes in structure including mismatched bases and simple methylation events to oxidized bases, intra- and interstrand DNA crosslinks, DNA double strand breaks and protein-DNA adducts. Pathways required for the repair of these lesions include mismatch repair, base excision repair, nucleotide excision repair, and the homology directed repair/Fanconi anemia pathway. Each of these pathways contributes to genetic stability, and mutations in genes encoding proteins involved in these pathways have been demonstrated to promote genetic instability and cancer. In fact, it has been suggested that all cancers display defects in DNA repair. It has also been demonstrated that the ability of cancer cells to repair therapeutically induced DNA damage impacts therapeutic efficacy. This has led to targeting DNA repair pathways and proteins to develop anti-cancer agents that will increase sensitivity to traditional chemotherapeutics. While initial studies languished and were plagued by a lack of specificity and a defined mechanism of action, more recent approaches to exploit synthetic lethal interaction and develop high affinity chemical inhibitors have proven considerably more effective. In this review we will highlight recent advances and discuss previous failures in targeting DNA repair to pave the way for future DNA repair targeted agents and their use in cancer therapy.
Collapse
Affiliation(s)
- Navnath S Gavande
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | | | - Hilary D Hinshaw
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Shadia I Jalal
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Catherine R Sears
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | | | - John J Turchi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States; NERx Biosciences, Indianapolis, IN 46202, United States; Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
17
|
Qi J, Dong Z, Liu J, Peery RC, Zhang S, Liu JY, Zhang JT. Effective Targeting of the Survivin Dimerization Interface with Small-Molecule Inhibitors. Cancer Res 2016; 76:453-62. [PMID: 26744521 DOI: 10.1158/0008-5472.can-15-1874] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/15/2015] [Indexed: 11/16/2022]
Abstract
Many oncoproteins are considered undruggable because they lack enzymatic activities. In this study, we present a small-molecule-based anticancer agent that acts by inhibiting dimerization of the oncoprotein survivin, thereby promoting its degradation along with spontaneous apoptosis in cancer cells. Through a combination of computational analysis of the dimerization interface and in silico screening, we identified one compound that induced proteasome-dependent survivin degradation. Analysis of a set of structural analogues led us to identify a lead compound (LQZ-7F), which was effective in blocking the survival of multiple cancer cell lines in a low micromolar concentration range. LQZ-7F induced proteasome-dependent survivin degradation, mitotic arrest, and apoptosis, and it blocked the growth of human tumors in mouse xenograft assays. In addition to providing preclinical proof of concept for a survivin-targeting anticancer agent, our work offers novel in silico screening strategies to therapeutically target homodimeric oncogenic proteins considered undruggable.
Collapse
Affiliation(s)
- Jing Qi
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Zizheng Dong
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jianguo Liu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Robert C Peery
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Shaobo Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jing-Yuan Liu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana. Department of Computer and Information Science, Indiana University-Purdue University at Indianapolis, Indiana.
| | - Jian-Ting Zhang
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana. IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
18
|
Lord T, Aitken RJ. Fertilization stimulates 8-hydroxy-2′-deoxyguanosine repair and antioxidant activity to prevent mutagenesis in the embryo. Dev Biol 2015; 406:1-13. [DOI: 10.1016/j.ydbio.2015.07.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/26/2015] [Accepted: 07/29/2015] [Indexed: 12/19/2022]
|
19
|
14-3-3σ confers cisplatin resistance in esophageal squamous cell carcinoma cells via regulating DNA repair molecules. Tumour Biol 2015; 37:2127-36. [PMID: 26346170 DOI: 10.1007/s13277-015-4018-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant type of esophageal cancer in Asia. Cisplatin is commonly used in chemoradiation for unresectable ESCC patients. However, the treatment efficacy is diminished in patients with established cisplatin resistance. To understand the mechanism leading to the development of cisplatin resistance in ESCC, we compared the proteomes from a cisplatin-resistant HKESC-2R cell line with its parental-sensitive counterpart HKESC-2 to identify key molecule involved in this process. Mass spectrometry analysis detected 14-3-3σ as the most abundant molecule expressed exclusively in HKESC-2R cells, while western blot result further validated it to be highly expressed in HKESC-2R cells when compared to HKESC-2 cells. Ectopic expression of 14-3-3σ increased cisplatin resistance in HKESC-2 cells, while its suppression sensitized SLMT-1 cells to cisplatin. Among the molecules involved in drug detoxification, drug transportation, and DNA repair, the examined DNA repair molecules HMGB1 and XPA were found to be highly expressed in HKESC-2R cells with high 14-3-3σ expression. Subsequent manipulation of 14-3-3σ by both overexpression and knockdown approaches concurrently altered the expression of HMGB1 and XPA. 14-3-3σ, HMGB1, and XPA were preferentially expressed in cisplatin-resistant SLMT-1 cells when compared to those more sensitive to cisplatin. In ESCC patients with poor response to cisplatin-based chemoradiation, their pre-treatment tumors expressed higher expression of HMGB1 than those with response to such treatment. In summary, our results demonstrate that 14-3-3σ induces cisplatin resistance in ESCC cells and that 14-3-3σ-mediated cisplatin resistance involves DNA repair molecules HMGB1 and XPA. Results from this study provide evidences for further work in researching the potential use of 14-3-3σ and DNA repair molecules HMGB1 and XPA as biomarkers and therapeutic targets for ESCC.
Collapse
|
20
|
Mishra AK, Dormi SS, Turchi AM, Woods DS, Turchi JJ. Chemical inhibitor targeting the replication protein A-DNA interaction increases the efficacy of Pt-based chemotherapy in lung and ovarian cancer. Biochem Pharmacol 2014; 93:25-33. [PMID: 25449597 DOI: 10.1016/j.bcp.2014.10.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 01/28/2023]
Abstract
Platinum-based chemotherapeutics exert their therapeutic efficacy via the formation of DNA adducts which interfere with DNA replication, transcription and cell division and ultimately induce cell death. Repair and tolerance of these Pt-DNA lesions by nucleotide excision repair (NER) and homologous recombination (HR) can substantially reduce the effectiveness of therapy. Inhibition of these repair pathways, therefore, holds the potential to sensitize cancer cells to Pt treatment and increase clinical efficacy. Replication Protein A (RPA) plays essential roles in both NER and HR, along with its role in DNA replication and DNA damage checkpoint activation. Each of these functions is, in part, mediated by RPA binding to single-stranded DNA (ssDNA). Here we report the synthesis and characterization of novel derivatives of RPA small molecule inhibitors and their activity in models of epithelial ovarian cancer (EOC) and non-small cell lung cancer (NSCLC). We have synthesized analogs of our previously reported RPA inhibitor TDRL-505 and determined the structure-activity relationships. These data led us to the identification of TDRL-551, which exhibited a greater than 2-fold increase in in vitro activity. TDRL-551 showed synergy with Pt in tissue culture models of EOC and in vivo efficacy, as a single agent and in combination with platinum, in a NSCLC xenograft model. These data demonstrate the utility of RPA inhibition in EOC and NSCLC and the potential in developing novel anticancer therapeutics that target RPA-DNA interactions.
Collapse
Affiliation(s)
- Akaash K Mishra
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Silvana S Dormi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alaina M Turchi
- NERx Biosciences Inc. 351W. 10th Street, Suite 510, Indianapolis, IN 46202, USA
| | - Derek S Woods
- NERx Biosciences Inc. 351W. 10th Street, Suite 510, Indianapolis, IN 46202, USA
| | - John J Turchi
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; NERx Biosciences Inc. 351W. 10th Street, Suite 510, Indianapolis, IN 46202, USA.
| |
Collapse
|
21
|
Abstract
![]()
Xeroderma
pigmentosum complementation group A (XPA) protein plays
a critical role in the repair of DNA damage via the nucleotide excision
repair (NER) pathway. XPA serves as a scaffold for NER, interacting
with several other NER proteins as well as the DNA substrate. The
critical importance of XPA is underscored by its association with
the most severe clinical phenotypes of the genetic disorder Xeroderma
pigmentosum. Many of these disease-associated mutations map to the
XPA98–219 DNA-binding domain (DBD) first reported
∼20 years ago. Although multiple solution NMR structures of
XPA98–219 have been determined, the molecular basis
for the interaction of this domain with DNA is only poorly characterized.
In this report, we demonstrate using a fluorescence anisotropy DNA-binding
assay that the previously reported XPA DBD binds DNA with substantially
weaker affinity than the full-length protein. In-depth analysis of
the XPA sequence suggested that the original DBD construct lacks critical
basic charge and helical elements at its C-terminus. Generation and
analysis of a series of C-terminal extensions beyond residue 219 yielded
a stable, soluble human XPA98–239 construct that
binds to a Y-shaped ssDNA–dsDNA junction and other substrates
with the same affinity as the full-length protein. Two-dimensional 15N–1H NMR suggested XPA98–239 contains the same globular core as XPA98–219 and
likely undergoes a conformational change upon binding DNA. Together,
our results demonstrate that the XPA DBD should be redefined and that
XPA98–239 is a suitable model to examine the DNA
binding activity of human XPA.
Collapse
Affiliation(s)
- Norie Sugitani
- Departments of Biochemistry and Chemistry, and Center for Structural Biology, Vanderbilt University , Nashville, Tennessee 37232-8725, United States
| | | | | | | |
Collapse
|
22
|
Allen WJ, Rizzo RC. Implementation of the Hungarian algorithm to account for ligand symmetry and similarity in structure-based design. J Chem Inf Model 2014; 54:518-29. [PMID: 24410429 PMCID: PMC3958141 DOI: 10.1021/ci400534h] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
![]()
False
negative docking outcomes for highly symmetric molecules
are a barrier to the accurate evaluation of docking programs, scoring
functions, and protocols. This work describes an implementation of
a symmetry-corrected root-mean-square deviation (RMSD) method into
the program DOCK based on the Hungarian algorithm for solving the
minimum assignment problem, which dynamically assigns atom correspondence
in molecules with symmetry. The algorithm adds only a trivial amount
of computation time to the RMSD calculations and is shown to increase
the reported overall docking success rate by approximately 5% when
tested over 1043 receptor–ligand systems. For some families
of protein systems the results are even more dramatic, with success
rate increases up to 16.7%. Several additional applications of the
method are also presented including as a pairwise similarity metric
to compare molecules during de novo design, as a scoring function
to rank-order virtual screening results, and for the analysis of trajectories
from molecular dynamics simulation. The new method, including source
code, is available to registered users of DOCK6 (http://dock.compbio.ucsf.edu).
Collapse
Affiliation(s)
- William J Allen
- Department of Applied Mathematics & Statistics, Stony Brook University , Stony Brook, New York 11794, United States
| | | |
Collapse
|
23
|
Small-molecule inhibitors of DNA damage-repair pathways: an approach to overcome tumor resistance to alkylating anticancer drugs. Future Med Chem 2012; 4:1093-111. [PMID: 22709253 DOI: 10.4155/fmc.12.58] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A major challenge in the future development of cancer therapeutics is the identification of biological targets and pathways, and the subsequent design of molecules to combat the drug-resistant cells hiding in virtually all cancers. This therapeutic approach is justified based upon the limited advances in cancer cures over the past 30 years, despite the development of many novel chemotherapies and earlier detection, which often fail due to drug resistance. Among the various targets to overcome tumor resistance are the DNA repair systems that can reverse the cytotoxicity of many clinically used DNA-damaging agents. Some progress has already been made but much remains to be done. We explore some components of the DNA-repair process, which are involved in repair of alkylation damage of DNA, as targets for the development of novel and effective molecules designed to improve the efficacy of existing anticancer drugs.
Collapse
|
24
|
Baughman BM, Jake Slavish P, DuBois RM, Boyd VA, White SW, Webb TR. Identification of influenza endonuclease inhibitors using a novel fluorescence polarization assay. ACS Chem Biol 2012; 7:526-34. [PMID: 22211528 DOI: 10.1021/cb200439z] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Influenza viruses have been responsible for the largest pandemics in the previous century. Although vaccination and prophylactic antiviral therapeutics are the primary defense against influenza virus, there is a pressing need to develop new antiviral agents to circumvent the limitations of current therapies. The endonuclease activity of the influenza virus PA(N) protein is essential for virus replication and is a promising target for novel anti-influenza drugs. To facilitate the discovery of endonuclease inhibitors, we have developed a high-throughput fluorescence polarization (FP) assay, utilizing a novel fluorescein-labeled compound (K(d) = 0.378 μM) and a PA(N) construct, to identify small molecules that bind to the PA(N) endonuclease active site. Several known 4-substituted 2,4-dioxobutanoic acid inhibitors with high and low affinities have been evaluated in this FP-based competitive binding assay, and there was a general correlation between binding and the reported inhibition of endonuclease activity. Additionally, we have demonstrated the utility of this assay for identifying endonuclease inhibitors in a small diverse targeted fragment library. These fragment hits were used to build a follow-up library that that led to new active compounds that demonstrate FP binding and anti-influenza activities in plaque inhibition assays. The assay offers significant advantages over previously reported assays and is suitable for high-throughput and fragment-based screening studies. Additionally the demonstration of the applicability of a mechanism-based "targeted fragment" library supports the general potential of this novel approach for other enzyme targets. These results serve as a sound foundation for the development of new therapeutic leads targeting influenza endonuclease.
Collapse
Affiliation(s)
- Brandi M. Baughman
- Integrated Program in Biomedical
Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | | | | | | | - Stephen W. White
- Integrated Program in Biomedical
Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Thomas R. Webb
- Integrated Program in Biomedical
Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
25
|
Jalal S, Earley JN, Turchi JJ. DNA repair: from genome maintenance to biomarker and therapeutic target. Clin Cancer Res 2011; 17:6973-84. [PMID: 21908578 DOI: 10.1158/1078-0432.ccr-11-0761] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A critical link exists between an individual's ability to repair cellular DNA damage and cancer development, progression, and response to therapy. Knowledge gained about the proteins involved and types of damage repaired by the individual DNA repair pathways has led to the development of a variety of assays aimed at determining an individual's DNA repair capacity. These assays and their use in the analysis of clinical samples have yielded useful though somewhat conflicting data. In this review article, we discuss the major DNA repair pathways, the proteins and genes required for each, assays used to analyze activity, and the relevant clinical studies to date. With the recent results from clinical trials targeting specific DNA repair proteins for the treatment of cancer, accurate, reproducible, and relevant analysis of DNA repair takes on an even greater significance. We highlight the strengths and limitations of these DNA repair studies and assays, with respect to the clinical assessment of DNA repair capacity to determine cancer development and response to therapy.
Collapse
Affiliation(s)
- Shadia Jalal
- Division of Hematology and Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
26
|
Role of eIF3a in regulating cisplatin sensitivity and in translational control of nucleotide excision repair of nasopharyngeal carcinoma. Oncogene 2011; 30:4814-23. [PMID: 21625209 PMCID: PMC3165083 DOI: 10.1038/onc.2011.189] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Translational control at the initiation step has been recognized as a major and important regulatory mechanism of gene expression. eIF3a, a putative subunit of eIF3 complex, has recently been shown to play an important role in regulating translation of a subset of mRNAs and found to correlate with prognosis of cancers. In this study, using nasopharyngeal carcinoma (NPC) cells as a model system we tested the hypothesis that eIF3a negatively regulates synthesis of nucleotide excision repair (NER) proteins and, thus, NER activities and cellular response to treatments with DNA damaging agents such as cisplatin. We found that a cisplatin-sensitive subclone S16 isolated from a NPC cell line CNE2 via limited dilution has increased eIF3a expression. Knocking down its expression in S16 cells increased cellular resistance to cisplatin, NER activity, and synthesis of NER proteins XPA, XPC, RAD23B, and RPA32. Altering eIF3a expression also changed cellular response to cisplatin and UV treatment in other NPC cell lines. Taken together, we conclude that eIF3a plays an important role in cisplatin response and NER activity of nasopharyngeal carcinomas by suppressing synthesis of NER proteins.
Collapse
|