1
|
Yang L, Sun Z. Role of APE1 in hepatocellular carcinoma and its prospects as a target in clinical settings (Review). Mol Clin Oncol 2024; 21:82. [PMID: 39301126 PMCID: PMC11411593 DOI: 10.3892/mco.2024.2780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
In recent years, the incidence of liver cancer has increased annually. However, current medical treatments for liver cancer are limited, and most patients have a high risk of recurrence after surgery. Therefore, the discovery and development of novel treatment targets for liver cancer is urgently needed. Apurinic/apyrimidinic endonuclease 1 (APE1) is a protein that has a DNA repair function and serves an important role in various physiological processes, including reduction-oxidation, cell proliferation and differentiation. The expression levels of APE1 are abnormally elevated in liver cancer cells, as ectopic expression of the APE1 gene has been reported, in addition to other abnormal signs, such as cell proliferation and migration. Therefore, it could be suggested that APE1 is an important indicator of hepatocellular carcinogenesis. APE1 may be used as a therapeutic target for tumors and proposed targeted therapy against abnormal APE1 expression could potentially inhibit the progression of tumors. The present review aimed to introduce the important role of APE1 in the physiological processes of tumor cells and the feasibility of using APE1 as a potential therapeutic target, providing a novel direction for the clinical treatment of liver cancer.
Collapse
Affiliation(s)
- Lei Yang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| | - Zhipeng Sun
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| |
Collapse
|
2
|
Siqueira PB, Rodrigues MMS, Amorim ĹSSD, Rodrigues JA, Oliveira MS, Fonseca AS, Pires BRB, Mencalha AL. The inhibitor of the redox activity of APE1/REF-1, APX2009, reduces the malignant phenotype of breast cancer cells. Braz J Med Biol Res 2024; 57:e13250. [PMID: 38808886 PMCID: PMC11136485 DOI: 10.1590/1414-431x2024e13250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/07/2024] [Indexed: 05/30/2024] Open
Abstract
Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/REF-1) is a multifunctional protein acting on cellular signaling pathways, including DNA repair and redox activities. APE1/REF-1 has emerged as a target for cancer therapy, and its role in breast cancer models would reveal new strategies for cancer therapy. APX2009 is a specific APE1/REF-1 redox inhibitor whose anticancer properties have not been described in breast cancer cells. Here, we investigated the effect of the APX2009 treatment in the breast cancer cell lines MDA-MB-231 and MCF-7. Breast cancer cell lines were cultured, and WST1 and colony formation assays were performed to evaluate cell proliferation. Annexin V-FITC/7-AAD and LDH-Glo™ assays were performed to evaluate cell death. The wound healing assay and Matrigel transwell assay were performed after APX2009 treatment to evaluate the cellular migration and invasion processes, respectively. Our findings demonstrated that APX2009 treatment decreased breast cancer cell proliferative, migratory, and invasive properties. Furthermore, it induced apoptosis in both cell lines. Our study is the first to show the effects of APX2009 treatment on apoptosis in a breast cancer cell. Therefore, this study suggested that APX2009 treatment is a promising anticancer molecule for breast cancer.
Collapse
Affiliation(s)
- P B Siqueira
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - M M S Rodrigues
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Ĺ S S de Amorim
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Laboratório de Alimentos Funcionais, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - J A Rodrigues
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - M S Oliveira
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - A S Fonseca
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - B R B Pires
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - A L Mencalha
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
3
|
Gampala S, Moon HR, Wireman R, Peil J, Kiran S, Mitchell DK, Brewster K, Mang H, Masters A, Bach C, Smith-Kinnamen W, Doud EH, Rai R, Mosley AL, Quinney SK, Clapp DW, Hamdouchi C, Wikel J, Zhang C, Han B, Georgiadis MM, Kelley MR, Fishel ML. New Ref-1/APE1 targeted inhibitors demonstrating improved potency for clinical applications in multiple cancer types. Pharmacol Res 2024; 201:107092. [PMID: 38311014 PMCID: PMC10962275 DOI: 10.1016/j.phrs.2024.107092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
AP endonuclease-1/Redox factor-1 (APE1/Ref-1 or Ref-1) is a multifunctional protein that is overexpressed in most aggressive cancers and impacts various cancer cell signaling pathways. Ref-1's redox activity plays a significant role in activating transcription factors (TFs) such as NFκB, HIF1α, STAT3 and AP-1, which are crucial contributors to the development of tumors and metastatic growth. Therefore, development of potent, selective inhibitors to target Ref-1 redox function is an appealing approach for therapeutic intervention. A first-generation compound, APX3330 successfully completed phase I clinical trial in adults with progressing solid tumors with favorable response rate, pharmacokinetics (PK), and minimal toxicity. These positive results prompted us to develop more potent analogs of APX3330 to effectively target Ref-1 in solid tumors. In this study, we present structure-activity relationship (SAR) identification and validation of lead compounds that exhibit a greater potency and a similar or better safety profile to APX3330. In order to triage and characterize the most potent and on-target second-generation Ref-1 redox inhibitors, we assayed for PK, mouse and human S9 fraction metabolic stability, in silico ADMET properties, ligand-based WaterLOGSY NMR measurements, pharmacodynamic markers, cell viability in multiple cancer cell types, and two distinct 3-dimensional (3D) cell killing assays (Tumor-Microenvironment on a Chip and 3D spheroid). To characterize the effects of Ref-1 inhibition in vivo, global proteomics was used following treatment with the top four analogs. This study identified and characterized more potent inhibitors of Ref-1 redox function (that outperformed APX3330 by 5-10-fold) with PK studies demonstrating efficacious doses for translation to clinic.
Collapse
Affiliation(s)
- Silpa Gampala
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, USA; Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Randall Wireman
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jacqueline Peil
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sonia Kiran
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dana K Mitchell
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kylee Brewster
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Henry Mang
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andi Masters
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Clinical Pharmacology Analytical Core, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Christine Bach
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Clinical Pharmacology Analytical Core, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Whitney Smith-Kinnamen
- Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma H Doud
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ratan Rai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amber L Mosley
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sara K Quinney
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - D Wade Clapp
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chafiq Hamdouchi
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - James Wikel
- Apexian Pharmaceuticals, Indianapolis, IN, USA
| | - Chi Zhang
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biohealth Informatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, USA; Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Millie M Georgiadis
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark R Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Melissa L Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
4
|
Mijit M, Kpenu E, Chowdhury NN, Gampala S, Wireman R, Liu S, Babb O, Georgiadis MM, Wan J, Fishel ML, Kelley MR. In vitro and In vivo evidence demonstrating chronic absence of Ref-1 Cysteine 65 impacts Ref-1 folding configuration, redox signaling, proliferation and metastasis in pancreatic cancer. Redox Biol 2024; 69:102977. [PMID: 38056311 PMCID: PMC10749280 DOI: 10.1016/j.redox.2023.102977] [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: 10/14/2023] [Revised: 11/13/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023] Open
Abstract
Ref-1/APE1 (Redox Effector/Apurinic Endonuclease 1) is a multifunctional enzyme that serves as a redox factor for several transcription factors (TFs), e.g., NF-kB, HIF-1α, which in an oxidized state fail to bind DNA. Conversion of these TFs to a reduced state serves to regulate various biological responses such as cell growth, inflammation, and cellular metabolism. The redox activity involves a thiol exchange reaction for which Cys65 (C65) serves as the nucleophile. Using CRISPR editing in human pancreatic ductal adenocarcinoma (PDAC) cells, we changed C65 to Ala (C65A) in Ref-1 to evaluate alteration of Ref-1 redox dynamics as well as chronic loss of Ref-1 redox activity on cell signaling pathways, specifically those regulated by NF-kB and HIF-1α. The redox activity of Ref-1 requires partial unfolding to expose C65, which is buried in the folded structure. Labeling of Ref-1 with polyethylene glycol-maleimide (PEGm) provides a readout of reduced Cys residues in Ref-1 and thereby an assessment of partial unfolding in Ref-1. In comparing Ref-1WT vs Ref-1C65A cell lines, we found an altered distribution of oxidized versus reduced states of Ref-1. Accordingly, activation of NF-kB and HIF-1α in Ref-1C65A lines was significantly lower compared to Ref-1WT lines. The bioinformatic data revealed significant downregulation of metabolic pathways including OXPHOS in Ref-1C65A expressing clones compared to Ref-1WT line. Ref-1C65A also demonstrated reduced cell proliferation and use of tricarboxylic acid (TCA) substrates compared to Ref-1WT lines. A subcutaneous as well as PDAC orthotopic in vivo model demonstrated a significant reduction in tumor size, weight, and growth in the Ref-1C65A lines compared to the Ref-1WT lines. Moreover, mice implanted with Ref-1C65A redox deficient cells demonstrate significantly reduced metastatic burden to liver and lung compared to mice implanted with Ref-1 redox proficient cells. These results from the current study provide direct evidence that the chronic absence of Cys65 in Ref-1 results in redox inactivity of the protein in human PDAC cells, and subsequent biological results confirm a critical involvement of Ref-1 redox signaling and tumorigenic phenotype.
Collapse
Affiliation(s)
- M Mijit
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E Kpenu
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N N Chowdhury
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - S Gampala
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Wireman
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Liu
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - O Babb
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M M Georgiadis
- Indiana University School of Medicine, Department of Biochemistry and Molecular Biology, Indianapolis, IN, USA
| | - J Wan
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - M L Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - M R Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University School of Medicine, Department of Biochemistry and Molecular Biology, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
Siqueira PB, de Sousa Rodrigues MM, de Amorim ÍSS, da Silva TG, da Silva Oliveira M, Rodrigues JA, de Souza da Fonseca A, Mencalha AL. The APE1/REF-1 and the hallmarks of cancer. Mol Biol Rep 2024; 51:47. [PMID: 38165468 DOI: 10.1007/s11033-023-08946-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/10/2023] [Indexed: 01/03/2024]
Abstract
APE1/REF-1 (apurinic/apyrimidinic endonuclease 1 / redox factor-1) is a protein with two domains, with endonuclease function and redox activity. Its main activity described is acting in DNA repair by base excision repair (BER) pathway, which restores DNA damage caused by oxidation, alkylation, and single-strand breaks. In contrast, the APE1 redox domain is responsible for regulating transcription factors, such as AP-1 (activating protein-1), NF-κB (Nuclear Factor kappa B), HIF-1α (Hypoxia-inducible factor 1-alpha), and STAT3 (Signal Transducers and Activators of Transcription 3). These factors are involved in physiological cellular processes, such as cell growth, inflammation, and angiogenesis, as well as in cancer. In human malignant tumors, APE1 overexpression is associated with lung, colon, ovaries, prostate, and breast cancer progression, more aggressive tumor phenotypes, and worse prognosis. In this review, we explore APE1 and its domain's role in cancer development processes, highlighting the role of APE1 in the hallmarks of cancer. We reviewed original articles and reviews from Pubmed related to APE1 and cancer and found that both domains of APE1/REF-1, but mainly its redox activity, are essential to cancer cells. This protein is often overexpressed in cancer, and its expression and activity are correlated to processes such as proliferation, invasion, inflammation, angiogenesis, and resistance to cell death. Therefore, APE1 participates in essential processes of cancer development. Then, the activity of APE1/REF-1 in these hallmarks suggests that targeting this protein could be a good therapeutic approach.
Collapse
Affiliation(s)
- Priscyanne Barreto Siqueira
- Departamento de Biofísica e Biometria, Laboratório de Biologia do Câncer, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil.
| | - Mariana Moreno de Sousa Rodrigues
- Departamento de Biofísica e Biometria, Laboratório de Biologia do Câncer, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil.
| | - Ísis Salviano Soares de Amorim
- Departamento de Biofísica e Biometria, Laboratório de Biologia do Câncer, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil
- Laboratório de Alimentos Funcionais, Universidade Federal do Rio de Janeiro, Instituto de Nutrição Josué de Castro, Rio de Janeiro, Brasil
| | - Thayssa Gomes da Silva
- Departamento de Biofísica e Biometria, Laboratório de Biofotônica, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil
| | - Matheus da Silva Oliveira
- Departamento de Biofísica e Biometria, Laboratório de Biologia do Câncer, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil
| | - Juliana Alves Rodrigues
- Departamento de Biofísica e Biometria, Laboratório de Biologia do Câncer, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil
| | - Adenilson de Souza da Fonseca
- Departamento de Biofísica e Biometria, Laboratório de Biofotônica, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil
| | - Andre Luiz Mencalha
- Departamento de Biofísica e Biometria, Laboratório de Biologia do Câncer, Universidade do Estado do Rio de Janeiro, Instituto de Biologia Roberto Alcântara Gomes, Rio de Janeiro, Brasil
| |
Collapse
|
6
|
Liu Y, Hao L, Dong Y, Dong BZ, Wang XL, Liu X, Hu ZX, Fang GC, Wang GY, Qin JX, Shi ZD, Pang K. Co-delivery of Siape1 and Melatonin by 125I-loaded PSMA-targeted Nanoparticles for the Treatment of Prostate Cancer. Recent Pat Anticancer Drug Discov 2024; 19:503-515. [PMID: 39044710 PMCID: PMC11348473 DOI: 10.2174/1574892818666230419081414] [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: 11/27/2022] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 07/25/2024]
Abstract
BACKGROUND Both apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) inhibition and melatonin suppress prostate cancer (PCa) growth. OBJECTIVE This study evaluated the therapeutic efficiency of self-assembled and prostate-specific membrane antigen (PSMA)-targeted nanocarrier loading 125I radioactive particles and encapsulating siRNA targeting APE1 (siAPE1) and melatonin for PCa. METHODS The linear polyarginine R12 polypeptide was prepared using Fmoc-Arg-Pbf-OH. The PSMA-targeted polymer was synthesized by conjugating azide-modified R12 peptide to PSMA monoclonal antibody (mAb). Before experiments, the PSMA-R12 nanocarrier was installed with melatonin and siAPE1, which were subsequently labeled by 125I radioactive particles. In vitro biocompatibility and cytotoxicity of nanocomposites were examined in LNCaP cells and in vivo biodistribution and pharmacokinetics were determined using PCa tumor-bearing mice. RESULTS PSMA-R12 nanocarrier was ~120 nm in size and was increased to ~150 nm by melatonin encapsulation. PSMA-R12 nanoparticles had efficient loading capacities of siAPE1, melatonin, and 125I particles. The co-delivery of melatonin and siAPE1 by PSMA-R12-125I showed synergistic effects on suppressing LNCaP cell proliferation and Bcl-2 expression and promoting cell apoptosis and caspase-3 expression. Pharmacokinetics analysis showed that Mel@PSMA-R12-125I particles had high uptake activity in the liver, spleen, kidney, intestine, and tumor, and were accumulated in the tumor sites within the first 8 h p.i., but was rapidly cleared from all the tested organs at 24 h p.i. Administration of nanoparticles to PCa tumors in vivo showed that Mel@PSMA-R12- 125I/siAPE1 had high efficiency in suppressing PCa tumor growth. CONCLUSION The PSMA-targeted nanocarrier encapsulating siAPE1 and melatonin is a promising therapeutic strategy for PCa and can provide a theoretical basis for patent applications.
Collapse
Affiliation(s)
- Ying Liu
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Life Sciences, Jiangsu Normal University, Jiangsu, China
| | - Yang Dong
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bing-Zheng Dong
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin-Lei Wang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
| | - Xing Liu
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
| | - Zheng-Xiang Hu
- Department of Graduate School, University of Jinzhou Medical University, Jinzhou, China
| | - Gao-Chuan Fang
- School of Life Sciences, Jiangsu Normal University, Jiangsu, China
| | - Guang-Yue Wang
- Department of Graduate School, University of Bengbu Medical College, Bengbu, China
| | - Jia-Xin Qin
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
| | - Zhen-Duo Shi
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Life Sciences, Jiangsu Normal University, Jiangsu, China
| | - Kun Pang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
7
|
Champion JD, Dodd KM, Lam HC, Alzahrani MAM, Seifan S, Rad E, Scourfield DO, Fishel ML, Calver BL, Ager A, Henske EP, Davies DM, Kelley MR, Tee AR. Drug Inhibition of Redox Factor-1 Restores Hypoxia-Driven Changes in Tuberous Sclerosis Complex 2 Deficient Cells. Cancers (Basel) 2022; 14:6195. [PMID: 36551683 PMCID: PMC9776744 DOI: 10.3390/cancers14246195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/17/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Therapies with the mechanistic target of rapamycin complex 1 (mTORC1) inhibitors are not fully curative for tuberous sclerosis complex (TSC) patients. Here, we propose that some mTORC1-independent disease facets of TSC involve signaling through redox factor-1 (Ref-1). Ref-1 possesses a redox signaling activity that stimulates the transcriptional activity of STAT3, NF-kB, and HIF-1α, which are involved in inflammation, proliferation, angiogenesis, and hypoxia, respectively. Here, we demonstrate that redox signaling through Ref-1 contributes to metabolic transformation and tumor growth in TSC cell model systems. In TSC2-deficient cells, the clinically viable Ref-1 inhibitor APX3330 was effective at blocking the hyperactivity of STAT3, NF-kB, and HIF-1α. While Ref-1 inhibitors do not inhibit mTORC1, they potently block cell invasion and vasculature mimicry. Of interest, we show that cell invasion and vasculature mimicry linked to Ref-1 redox signaling are not blocked by mTORC1 inhibitors. Metabolic profiling revealed that Ref-1 inhibitors alter metabolites associated with the glutathione antioxidant pathway as well as metabolites that are heavily dysregulated in TSC2-deficient cells involved in redox homeostasis. Therefore, this work presents Ref-1 and associated redox-regulated transcription factors such as STAT3, NF-kB, and HIF-1α as potential therapeutic targets to treat TSC, where targeting these components would likely have additional benefits compared to using mTORC1 inhibitors alone.
Collapse
Affiliation(s)
- Jesse D. Champion
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Kayleigh M. Dodd
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Hilaire C. Lam
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Sara Seifan
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Ellie Rad
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | | | - Melissa L. Fishel
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brian L. Calver
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Ann Ager
- Division of Infection and Immunity, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Elizabeth P. Henske
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David Mark Davies
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
- Department of Oncology, South West Wales Cancer Centre, Singleton Hospital, Swansea SA2 8QA, UK
| | - Mark R. Kelley
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew R. Tee
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
8
|
Hai Z, Jia Q. Apurinic/apyrimidinic endonuclease 1 mRNA level in peripheral blood neutrophils is associated with asthma. INVESTIGACIÓN CLÍNICA 2022. [DOI: 10.54817/ic.v63n4a02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunc-tional key protein. Recent studies suggest APE1 is closely associated with in-flammatory response, but its role in asthma remains unknown. We recruited 116 patients with asthma, including 50 with severe asthma (NSA) and 66 with non-severe asthma (SA), and 140 controls. Serum APE1 was detected using the ELISA method. APE1 mRNA in peripheral blood neutrophils and eosinophils were detected using real-time PCR assays. Compared to healthy controls, we observed significant elevations of serum APE1 mRNA levels in peripheral neu-trophils (~1.75 folds increase, p<0.05) and eosinophils (~2.2 folds increase, p<0.05) in patients with asthma. The peripheral blood neutrophil APE1 mRNA can distinguish asthmatic patients from healthy controls with the area under the curve (AUC) 0.893 and a 95% confidence interval (CI) 0.847-0.938 (p < 0.001). Also the APE1 mRNA can identify severe asthma from non-severe asth-ma (AUC 0.759, 95% CI, 0.674-0.846; p < 0.001). However, The serum APE1 and eosinophil mRNA levels did not correlate with asthma incidence and sever-ity. Our finding confirms the association between APE1 and asthma and sug-gests that peripheral blood neutrophil APE1 mRNA may be used as a marker for this condition.
Collapse
Affiliation(s)
- Zhou Hai
- Department of Respiration, Shidong Hospital of Yangpu District, No. 999, Shi Guang Rd, Yangpu District, Shanghai 200438, China
| | - Qin Jia
- Department of Respiration, Shidong Hospital of Yangpu District, No. 999, Shi Guang Rd, Yangpu District, Shanghai 200438, China
| |
Collapse
|
9
|
Alhazmi R, Tong S, Darwish S, Khanjani E, Khungar B, Chawla S, Zheng Z, Chamberlin R, Parang K, Yang S. Bis-Cinnamamide Derivatives as APE/Ref-1 Inhibitors for the Treatment of Human Melanoma. Molecules 2022; 27:2672. [PMID: 35566022 PMCID: PMC9103902 DOI: 10.3390/molecules27092672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 12/04/2022] Open
Abstract
Human malignant melanoma exhibits imbalances in redox status, leading to activation of many redox-sensitive signaling pathways. APE/Ref-1 is a multifunctional protein that serves as a redox chaperone that regulates many nuclear transcription factors and is an important mechanism in cancer cell survival of oxidative stress. Previous studies showed that APE/Ref-1 is a potential druggable target for melanoma therapy. In this study, we synthesized a novel APE/Ref-1 inhibitor, bis-cinnamoyl-1,12-dodecamethylenediamine (2). In a xenograft mouse model, compound 2 treatment (5 mg/kg) significantly inhibited tumor growth compared to the control group, with no significant systemic toxicity observed. We further synthesized compound 2 analogs to determine the structure-activity relationship based on their anti-melanoma activities. Among those, 4-hydroxyphenyl derivative (11) exhibited potent anti-melanoma activities and improved water solubility compared to its parental compound 2. The IC50 of compound 11 was found to be less than 0.1 μM. Compared to other known APE/Ref-1 inhibitors, compound 11 exhibited increased potency in inhibiting melanoma proliferation. As determined by luciferase reporter analyses, compound 2 was shown to effectively inhibit H2O2-activated AP-1 transcription activities. Targeting APE/Ref-1-mediated signaling using pharmaceutical inhibitors is a novel and effective strategy for melanoma treatment with potentially high impact.
Collapse
Affiliation(s)
- Razan Alhazmi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Shirley Tong
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Shaban Darwish
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Elina Khanjani
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Bharti Khungar
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Swati Chawla
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Zhonghui Zheng
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA; (Z.Z.); (R.C.)
| | - Richard Chamberlin
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA; (Z.Z.); (R.C.)
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (R.A.); (S.T.); (S.D.); (E.K.); (B.K.); (S.C.)
| | - Sun Yang
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
10
|
Mijit M, Wireman R, Armstrong L, Gampala S, Hassan Z, Schneeweis C, Schneider G, Zhang C, Fishel ML, Kelley MR. RelA Is an Essential Target for Enhancing Cellular Responses to the DNA Repair/Ref-1 Redox Signaling Protein and Restoring Perturbated Cellular Redox Homeostasis in Mouse PDAC Cells. Front Oncol 2022; 12:826617. [PMID: 35402225 PMCID: PMC8988139 DOI: 10.3389/fonc.2022.826617] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/22/2022] [Indexed: 11/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers with a poor response to current treatment regimens. The multifunctional DNA repair-redox signaling protein Ref-1 has a redox signaling function that activates several transcriptional factors (TFs) including NF-κB (RelA), STAT3, AP-1. These have been implicated in signaling in PDAC and associated with cancer progression and therapy resistance. Numerous studies have shown a role for RelA in PDAC inflammatory responses and therapy resistance, little is known as to how these inflammatory responses are modulated through Ref-1 redox signaling pathways during pancreatic pathogenesis. RelA and STAT3 are two major targets of Ref-1 and are important in PDAC pathogenesis. To decipher the mechanistic role of RelA in response to Ref-1 inhibition, we used PDAC cells (KC3590) from a genetically engineered Kras G12D-driven mouse model that also is functionally deficient for RelA (Parent/Vector) or KC3590 cells with fully functional RelA added back (clone 13; C13). We demonstrated that RelA deficient cells are more resistant to Ref-1 redox inhibitors APX3330, APX2009, and APX2014, and their sensitivity is restored in the RelA proficient cells. Knockdown of STAT3 did not change cellular sensitivity to Ref-1 redox inhibitors in either cell type. Gene expression analysis demonstrated that Ref-1 inhibitors significantly decreased IL-8, FOSB, and c-Jun when functional RelA is present. We also demonstrated that PRDX1, a known Ref-1 redox modulator, contributes to Ref-1 inhibitor cellular response. Knockdown of PRDX1 when functional RelA is present resulted in dramatically increased PDAC killing in response to Ref-1 inhibitors. The enhanced cell killing was not due to increased intracellular ROS production. Although Ref-1 inhibition decreased the NADP/NADPH ratio in the cells, the addition of PRDX1 knockdown did not further this redox imbalance. This data suggests that the mechanism of cell killing following Ref-1 inhibition is at least partially mediated through RelA and not STAT3. Further imbalancing of the redox signaling through disruption of the PRDX1-Ref-1 interaction may have therapeutic implications. Our data further support a pivotal role of RelA in mediating Ref-1 redox signaling in PDAC cells with the Kras G12D genotype and provide novel therapeutic strategies to combat PDAC drug resistance.
Collapse
Affiliation(s)
- Mahmut Mijit
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Randall Wireman
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lee Armstrong
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Silpa Gampala
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zonera Hassan
- Department of Clinic and Polyclinic for Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christian Schneeweis
- Department of Clinic and Polyclinic for Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Guenter Schneider
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biohealth Informatics, Indiana University-Purdue University (IUPUI), Indianapolis, IN, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Melissa L. Fishel
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mark R. Kelley
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
11
|
Inhibition of APE1/Ref-1 for Neovascular Eye Diseases: From Biology to Therapy. Int J Mol Sci 2021; 22:ijms221910279. [PMID: 34638620 PMCID: PMC8508814 DOI: 10.3390/ijms221910279] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
Proliferative diabetic retinopathy (PDR), neovascular age-related macular degeneration (nvAMD), retinopathy of prematurity (ROP) and other eye diseases are characterized by retinal and/or choroidal neovascularization, ultimately causing vision loss in millions of people worldwide. nvAMD and PDR are associated with aging and the number of those affected is expected to increase as the global median age and life expectancy continue to rise. With this increase in prevalence, the development of novel, orally bioavailable therapies for neovascular eye diseases that target multiple pathways is critical, since current anti-vascular endothelial growth factor (VEGF) treatments, delivered by intravitreal injection, are accompanied with tachyphylaxis, a high treatment burden and risk of complications. One potential target is apurinic/apyrimidinic endonuclease 1/reduction-oxidation factor 1 (APE1/Ref-1). The multifunctional protein APE1/Ref-1 may be targeted via inhibitors of its redox-regulating transcription factor activation activity to modulate angiogenesis, inflammation, oxidative stress response and cell cycle in neovascular eye disease; these inhibitors also have neuroprotective effects in other tissues. An APE1/Ref-1 small molecule inhibitor is already in clinical trials for cancer, PDR and diabetic macular edema. Efforts to develop further inhibitors are underway. APE1/Ref-1 is a novel candidate for therapeutically targeting neovascular eye diseases and alleviating the burden associated with anti-VEGF intravitreal injections.
Collapse
|
12
|
Fleming AM, Burrows CJ. Oxidative stress-mediated epigenetic regulation by G-quadruplexes. NAR Cancer 2021; 3:zcab038. [PMID: 34541539 PMCID: PMC8445369 DOI: 10.1093/narcan/zcab038] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Many cancer-associated genes are regulated by guanine (G)-rich sequences that are capable of refolding from the canonical duplex structure to an intrastrand G-quadruplex. These same sequences are sensitive to oxidative damage that is repaired by the base excision repair glycosylases OGG1 and NEIL1–3. We describe studies indicating that oxidation of a guanosine base in a gene promoter G-quadruplex can lead to up- and downregulation of gene expression that is location dependent and involves the base excision repair pathway in which the first intermediate, an apurinic (AP) site, plays a key role mediated by AP endonuclease 1 (APE1/REF1). The nuclease activity of APE1 is paused at a G-quadruplex, while the REF1 capacity of this protein engages activating transcription factors such as HIF-1α, AP-1 and p53. The mechanism has been probed by in vitro biophysical studies, whole-genome approaches and reporter plasmids in cellulo. Replacement of promoter elements by a G-quadruplex sequence usually led to upregulation, but depending on the strand and precise location, examples of downregulation were also found. The impact of oxidative stress-mediated lesions in the G-rich sequence enhanced the effect, whether it was positive or negative.
Collapse
Affiliation(s)
- Aaron M Fleming
- Department of Chemistry, University of Utah, 315 S. 1400 East, Salt Lake City, UT 84112-0850, USA
| | - Cynthia J Burrows
- Department of Chemistry, University of Utah, 315 S. 1400 East, Salt Lake City, UT 84112-0850, USA
| |
Collapse
|
13
|
Gampala S, Shah F, Lu X, Moon HR, Babb O, Umesh Ganesh N, Sandusky G, Hulsey E, Armstrong L, Mosely AL, Han B, Ivan M, Yeh JRJ, Kelley MR, Zhang C, Fishel ML. Ref-1 redox activity alters cancer cell metabolism in pancreatic cancer: exploiting this novel finding as a potential target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:251. [PMID: 34376225 PMCID: PMC8353735 DOI: 10.1186/s13046-021-02046-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 07/18/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic cancer is a complex disease with a desmoplastic stroma, extreme hypoxia, and inherent resistance to therapy. Understanding the signaling and adaptive response of such an aggressive cancer is key to making advances in therapeutic efficacy. Redox factor-1 (Ref-1), a redox signaling protein, regulates the conversion of several transcription factors (TFs), including HIF-1α, STAT3 and NFκB from an oxidized to reduced state leading to enhancement of their DNA binding. In our previously published work, knockdown of Ref-1 under normoxia resulted in altered gene expression patterns on pathways including EIF2, protein kinase A, and mTOR. In this study, single cell RNA sequencing (scRNA-seq) and proteomics were used to explore the effects of Ref-1 on metabolic pathways under hypoxia. METHODS scRNA-seq comparing pancreatic cancer cells expressing less than 20% of the Ref-1 protein was analyzed using left truncated mixture Gaussian model and validated using proteomics and qRT-PCR. The identified Ref-1's role in mitochondrial function was confirmed using mitochondrial function assays, qRT-PCR, western blotting and NADP assay. Further, the effect of Ref-1 redox function inhibition against pancreatic cancer metabolism was assayed using 3D co-culture in vitro and xenograft studies in vivo. RESULTS Distinct transcriptional variation in central metabolism, cell cycle, apoptosis, immune response, and genes downstream of a series of signaling pathways and transcriptional regulatory factors were identified in Ref-1 knockdown vs Scrambled control from the scRNA-seq data. Mitochondrial DEG subsets downregulated with Ref-1 knockdown were significantly reduced following Ref-1 redox inhibition and more dramatically in combination with Devimistat in vitro. Mitochondrial function assays demonstrated that Ref-1 knockdown and Ref-1 redox signaling inhibition decreased utilization of TCA cycle substrates and slowed the growth of pancreatic cancer co-culture spheroids. In Ref-1 knockdown cells, a higher flux rate of NADP + consuming reactions was observed suggesting the less availability of NADP + and a higher level of oxidative stress in these cells. In vivo xenograft studies demonstrated that tumor reduction was potent with Ref-1 redox inhibitor similar to Devimistat. CONCLUSION Ref-1 redox signaling inhibition conclusively alters cancer cell metabolism by causing TCA cycle dysfunction while also reducing the pancreatic tumor growth in vitro as well as in vivo.
Collapse
Affiliation(s)
- Silpa Gampala
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaoyu Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Biohealth Informatics, IUPUI, Indianapolis, IN, 46202, USA
| | - Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Olivia Babb
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nikkitha Umesh Ganesh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, 02115, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - George Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine , Indianapolis, IN, 46202, USA.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W Walnut St. R4-321, Indianapolis, IN, 46202, USA
| | - Emily Hulsey
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine , Indianapolis, IN, 46202, USA
| | - Lee Armstrong
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Amber L Mosely
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W Walnut St. R4-321, Indianapolis, IN, 46202, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47906, USA.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, 47906, USA
| | - Mircea Ivan
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W Walnut St. R4-321, Indianapolis, IN, 46202, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jing-Ruey Joanna Yeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, 02115, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Mark R Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W Walnut St. R4-321, Indianapolis, IN, 46202, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Biohealth Informatics, IUPUI, Indianapolis, IN, 46202, USA. .,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W Walnut St. R4-321, Indianapolis, IN, 46202, USA.
| | - Melissa L Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W Walnut St. R4-321, Indianapolis, IN, 46202, USA. .,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
14
|
Gampala S, Shah F, Zhang C, Rhodes SD, Babb O, Grimard M, Wireman RS, Rad E, Calver B, Bai RY, Staedtke V, Hulsey EL, Saadatzadeh MR, Pollok KE, Tong Y, Smith AE, Clapp DW, Tee AR, Kelley MR, Fishel ML. Exploring transcriptional regulators Ref-1 and STAT3 as therapeutic targets in malignant peripheral nerve sheath tumours. Br J Cancer 2021; 124:1566-1580. [PMID: 33658640 PMCID: PMC8076291 DOI: 10.1038/s41416-021-01270-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 12/03/2020] [Accepted: 01/05/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND MPNST is a rare soft-tissue sarcoma that can arise from patients with NF1. Existing chemotherapeutic and targeted agents have been unsuccessful in MPNST treatment, and recent findings implicate STAT3 and HIF1-α in driving MPNST. The DNA-binding and transcriptional activity of both STAT3 and HIF1-α is regulated by Redox factor-1 (Ref-1) redox function. A first-generation Ref-1 inhibitor, APX3330, is being tested in cancer clinical trials and could be applied to MPNST. METHODS We characterised Ref-1 and p-STAT3 expression in various MPNST models. Tumour growth, as well as biomarkers of apoptosis and signalling pathways, were measured by qPCR and western blot following treatment with inhibitors of Ref-1 or STAT3. RESULTS MPNSTs from Nf1-Arfflox/floxPostnCre mice exhibit significantly increased positivity of p-STAT3 and Ref-1 expression when malignant transformation occurs. Inhibition of Ref-1 or STAT3 impairs MPNST growth in vitro and in vivo and induces apoptosis. Genes highly expressed in MPNST patients are downregulated following inhibition of Ref-1 or STAT3. Several biomarkers downstream of Ref-1 or STAT3 were also downregulated following Ref-1 or STAT3 inhibition. CONCLUSIONS Our findings implicate a unique therapeutic approach to target important MPNST signalling nodes in sarcomas using new first-in-class small molecules for potential translation to the clinic.
Collapse
Affiliation(s)
- Silpa Gampala
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, USA.,Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Steven D Rhodes
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Olivia Babb
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Michelle Grimard
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Randall S Wireman
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Ellie Rad
- Division of Cancer and Genetics, Cardiff University, Cardiff, Wales, UK
| | - Brian Calver
- Division of Cancer and Genetics, Cardiff University, Cardiff, Wales, UK
| | - Ren-Yuan Bai
- Neurosurgery and Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Verena Staedtke
- Neurosurgery and Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Emily L Hulsey
- Department of Pathology and Laboratory Medicine, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - M Reza Saadatzadeh
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Karen E Pollok
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Yan Tong
- Department of Biostatistics and Data Management, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Abbi E Smith
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - D Wade Clapp
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Andrew R Tee
- Division of Cancer and Genetics, Cardiff University, Cardiff, Wales, UK
| | - Mark R Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Melissa L Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
15
|
Greulich BM, Plotnik JP, Jerde TJ, Hollenhorst PC. Toll-like receptor 4 signaling activates ERG function in prostate cancer and provides a therapeutic target. NAR Cancer 2021; 3:zcaa046. [PMID: 33554122 PMCID: PMC7848947 DOI: 10.1093/narcan/zcaa046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
The TMPRSS2–ERG gene fusion and subsequent overexpression of the ERG transcription factor occurs in ∼50% of prostate tumors, making it the most common abnormality of the prostate cancer genome. While ERG has been shown to drive tumor progression and cancer-related phenotypes, as a transcription factor it is difficult to target therapeutically. Using a genetic screen, we identified the toll-like receptor 4 (TLR4) signaling pathway as important for ERG function in prostate cells. Our data confirm previous reports that ERG can transcriptionally activate TLR4 gene expression; however, using a constitutively active ERG mutant, we demonstrate that the critical function of TLR4 signaling is upstream, promoting ERG phosphorylation at serine 96 and ERG transcriptional activation. The TLR4 inhibitor, TAK-242, attenuated ERG-mediated migration, clonogenic survival, target gene activation and tumor growth. Together these data indicate a mechanistic basis for inhibition of TLR4 signaling as a treatment for ERG-positive prostate cancer.
Collapse
Affiliation(s)
- Benjamin M Greulich
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Joshua P Plotnik
- Biology Department, Indiana University, Bloomington, IN 47405, USA
| | - Travis J Jerde
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA
| |
Collapse
|
16
|
Caston RA, Gampala S, Armstrong L, Messmann RA, Fishel ML, Kelley MR. The multifunctional APE1 DNA repair-redox signaling protein as a drug target in human disease. Drug Discov Today 2021; 26:218-228. [PMID: 33148489 PMCID: PMC7855940 DOI: 10.1016/j.drudis.2020.10.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/27/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Apurinic/apyrimidinic (AP) endonuclease-reduction/oxidation factor 1 (APE1/Ref-1, also called APE1) is a multifunctional enzyme with crucial roles in DNA repair and reduction/oxidation (redox) signaling. APE1 was originally described as an endonuclease in the Base Excision Repair (BER) pathway. Further study revealed it to be a redox signaling hub regulating critical transcription factors (TFs). Although a significant amount of focus has been on the role of APE1 in cancer, recent findings support APE1 as a target in other indications, including ocular diseases [diabetic retinopathy (DR), diabetic macular edema (DME), and age-related macular degeneration (AMD)], inflammatory bowel disease (IBD) and others, where APE1 regulation of crucial TFs impacts important pathways in these diseases. The central responsibilities of APE1 in DNA repair and redox signaling make it an attractive therapeutic target for cancer and other diseases.
Collapse
Affiliation(s)
- Rachel A Caston
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | - Silpa Gampala
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | - Lee Armstrong
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | | | - Melissa L Fishel
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | - Mark R Kelley
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA.
| |
Collapse
|
17
|
Caston RA, Shah F, Starcher CL, Wireman R, Babb O, Grimard M, McGeown J, Armstrong L, Tong Y, Pili R, Rupert J, Zimmers TA, Elmi AN, Pollok KE, Motea EA, Kelley MR, Fishel ML. Combined inhibition of Ref-1 and STAT3 leads to synergistic tumour inhibition in multiple cancers using 3D and in vivo tumour co-culture models. J Cell Mol Med 2021; 25:784-800. [PMID: 33274592 PMCID: PMC7812272 DOI: 10.1111/jcmm.16132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/19/2020] [Accepted: 11/09/2020] [Indexed: 12/26/2022] Open
Abstract
With a plethora of molecularly targeted agents under investigation in cancer, a clear need exists to understand which pathways can be targeted simultaneously with multiple agents to elicit a maximal killing effect on the tumour. Combination therapy provides the most promise in difficult to treat cancers such as pancreatic. Ref-1 is a multifunctional protein with a role in redox signalling that activates transcription factors such as NF-κB, AP-1, HIF-1α and STAT3. Formerly, we have demonstrated that dual targeting of Ref-1 (redox factor-1) and STAT3 is synergistic and decreases cell viability in pancreatic cancer cells. Data presented here extensively expands upon this work and provides further insights into the relationship of STAT3 and Ref-1 in multiple cancer types. Using targeted small molecule inhibitors, Ref-1 redox signalling was blocked along with STAT3 activation, and tumour growth evaluated in the presence and absence of the relevant tumour microenvironment. Our study utilized qPCR, cytotoxicity and in vivo analysis of tumour and cancer-associated fibroblasts (CAF) response to determine the synergy of Ref-1 and STAT3 inhibitors. Overall, pancreatic tumours grown in the presence of CAFs were sensitized to the combination of STAT3 and Ref-1 inhibition in vivo. In vitro bladder and pancreatic cancer demonstrated the most synergistic responses. By disabling both of these important pathways, this combination therapy has the capacity to hinder crosstalk between the tumour and its microenvironment, leading to improved tumour response.
Collapse
Affiliation(s)
- Rachel A. Caston
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Colton L. Starcher
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
| | - Randall Wireman
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Olivia Babb
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Michelle Grimard
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Jack McGeown
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Lee Armstrong
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Yan Tong
- Department of BiostatisticsIndiana University School of MedicineIndianapolisINUSA
| | - Roberto Pili
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Department of UrologyIndiana University School of MedicineIndianapolisINUSA
- Department of Hematology and OncologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Joseph Rupert
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
- Department of SurgeryIndiana University School of MedicineIndianapolisINUSA
| | - Teresa A. Zimmers
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
- Department of SurgeryIndiana University School of MedicineIndianapolisINUSA
- Richard L. Roudebush Veterans Administration Medical CenterIndianapolisINUSA
| | - Adily N. Elmi
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
| | - Karen E. Pollok
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Edward A. Motea
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Mark R. Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| | - Melissa L. Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndiana University School of MedicineIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
18
|
Felix FA, da Silva LP, Lopes MLDDS, Sobral APV, Freitas RDA, de Souza LB, Barboza CAG. DNA base excision repair and nucleotide excision repair proteins in malignant salivary gland tumors. Arch Oral Biol 2020; 121:104987. [PMID: 33202356 DOI: 10.1016/j.archoralbio.2020.104987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To analyze the immunohistochemical expression of the base excision repair (BER) proteins apurinic/apyrimidinic endonuclease 1 (APE1) and X-ray repair cross-complementing protein 1 (XRCC1) and nucleotide excision repair (NER) protein xeroderma pigmentosum group F (XPF) in malignant salivary gland tumors (MSGTs). DESIGN Sixty-two cases of MSGTs were selected, including 14 acinic cell carcinomas (AcCC), 15 polymorphous adenocarcinomas (PAC), 16 adenoid cystic carcinomas (ACC), and 17 mucoepidermoid carcinomas (MEC). The specimens were submitted to quantitative immunohistochemical analysis. RESULTS All MSGTs exhibited nuclear or nucleo-cytoplasmic immunostaining of APE1, XRCC1 and XPF, with a high percentage of positive cells (median = 78.31, 70.48 and 75.46, respectively). XRCC1 expression was higher in PAC compared to MEC (p = 0.032). Nuclear APE1 immunostaining was significantly higher than nucleo-cytoplasmic expression in the selected MSGTs (p < 0.0001). APE1 expression was significantly associated with T1-T2 tumors in ACC (p = 0.006). Increased expression of XPF was associated with age older than 60 years in MEC (p = 0.015) and with ACC involving the minor salivary gland (p = 0.012), while a lower expression was found in AcCC and ACC patients treated by surgery combined with adjuvant therapy (p = 0.036 and p = 0.020, respectively). Low expression of XRCC1 in the nucleus (p = 0.028) and concomitant expression of this protein in the nucleus/cytoplasm were associated with a lower overall 5-year survival rate (p = 0.017). CONCLUSIONS This study showed that BER and NER proteins evaluated are highly expressed in the MSGTs studied, indicating mechanisms of genotoxic control in these tumors. In addition, the dysregulation of XRCC1 expression was a prognostic predictor in MSGTs analyzed.
Collapse
Affiliation(s)
- Fernanda Aragão Felix
- Postgraduate Program in Dental Science, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | | | | | - Roseana de Almeida Freitas
- Postgraduate Program in Dental Science, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Lélia Batista de Souza
- Postgraduate Program in Dental Science, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Carlos Augusto Galvão Barboza
- Postgraduate Program in Dental Science, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
19
|
Franko A, Berti L, Guirguis A, Hennenlotter J, Wagner R, Scharpf MO, de Angelis MH, Wißmiller K, Lickert H, Stenzl A, Birkenfeld AL, Peter A, Häring HU, Lutz SZ, Heni M. Characterization of Hormone-Dependent Pathways in Six Human Prostate-Cancer Cell Lines: A Gene-Expression Study. Genes (Basel) 2020; 11:E1174. [PMID: 33036464 PMCID: PMC7599530 DOI: 10.3390/genes11101174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa), the most incident cancer in men, is tightly regulated by endocrine signals. A number of different PCa cell lines are commonly used for in vitro experiments, but these are of diverse origin, and have very different cell-proliferation rates and hormone-response capacities. By analyzing the gene-expression pattern of main hormone pathways, we systematically compared six PCa cell lines and parental primary cells. We compared these cell lines (i) with each other and (ii) with PCa tissue samples from 11 patients. We found major differences in the gene-expression levels of androgen, insulin, estrogen, and oxysterol signaling between PCa tissue and cell lines, and between different cell lines. Our systematic characterization gives researchers a solid basis to choose the appropriate PCa cell model for the hormone pathway of interest.
Collapse
Affiliation(s)
- Andras Franko
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Lucia Berti
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Alke Guirguis
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (A.S.)
| | - Robert Wagner
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Marcus O. Scharpf
- Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Martin Hrabĕ de Angelis
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Katharina Wißmiller
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany;
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Medicine, Technical University of Munich, 81675 München, Germany
| | - Heiko Lickert
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany;
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Medicine, Technical University of Munich, 81675 München, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (A.S.)
| | - Andreas L. Birkenfeld
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
| | - Stefan Z. Lutz
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, 72116 Mössingen, Germany
| | - Martin Heni
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (R.W.); (A.L.B.); (H.-U.H.); (S.Z.L.); (M.H.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; (M.H.d.A.); (H.L.)
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
20
|
Franko A, Berti L, Hennenlotter J, Rausch S, Scharpf MO, de Angelis MH, Stenzl A, Birkenfeld AL, Peter A, Lutz SZ, Häring HU, Heni M. Transcript Levels of Aldo-Keto Reductase Family 1 Subfamily C (AKR1C) Are Increased in Prostate Tissue of Patients with Type 2 Diabetes. J Pers Med 2020; 10:jpm10030124. [PMID: 32932589 PMCID: PMC7564141 DOI: 10.3390/jpm10030124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Aldo-keto reductase family 1 (AKR1) enzymes play a crucial role in diabetic complications. Since type 2 diabetes (T2D) is associated with cancer progression, we investigated the impact of diabetes on AKR1 gene expression in the context of prostate cancer (PCa) development. In this study, we analyzed benign (BEN) prostate and PCa tissue of patients with and without T2D. Furthermore, to replicate hyperglycemia in vitro, we treated the prostate adenocarcinoma cell line PC3 with increasing glucose concentrations. Gene expression was quantified using real-time qPCR. In the prostate tissue of patients with T2D, AKR1C1 and AKR1C2 transcripts were higher compared to samples of patients without diabetes. In PC3 cells, high glucose treatment induced the gene expression levels of AKR1C1, C2, and C3. Furthermore, both in human tissue and in PC3 cells, the transcript levels of AKR1C1, C2, and C3 showed positive associations with oncogenes, which are involved in proliferation processes and HIF1α and NFκB pathways. These results indicate that in the prostate glands of patients with T2D, hyperglycemia could play a pivotal role by inducing the expression of AKR1C1, C2, and C3. The higher transcript level of AKR1C was furthermore associated with upregulated HIF1α and NFκB pathways, which are major drivers of PCa carcinogenesis.
Collapse
Affiliation(s)
- Andras Franko
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Lucia Berti
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (S.R.); (A.S.)
| | - Steffen Rausch
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (S.R.); (A.S.)
| | - Marcus O. Scharpf
- Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Martin Hrabĕ de Angelis
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (S.R.); (A.S.)
| | - Andreas L. Birkenfeld
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (A.P.)
| | - Stefan Z. Lutz
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, 72116 Mössingen, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Martin Heni
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (A.P.)
- Correspondence: ; Tel.: +49-7071-29-82714
| |
Collapse
|
21
|
Franko A, Shao Y, Heni M, Hennenlotter J, Hoene M, Hu C, Liu X, Zhao X, Wang Q, Birkenfeld AL, Todenhöfer T, Stenzl A, Peter A, Häring HU, Lehmann R, Xu G, Lutz SZ. Human Prostate Cancer is Characterized by an Increase in Urea Cycle Metabolites. Cancers (Basel) 2020; 12:E1814. [PMID: 32640711 PMCID: PMC7408908 DOI: 10.3390/cancers12071814] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/18/2022] Open
Abstract
Despite it being the most common incident of cancer among men, the pathophysiological mechanisms contributing to prostate cancer (PCa) are still poorly understood. Altered mitochondrial metabolism is postulated to play a role in the development of PCa. To determine the key metabolites (which included mitochondrial oncometabolites), benign prostatic and cancer tissues of patients with PCa were analyzed using capillary electrophoresis and liquid chromatography coupled with mass spectrometry. Gene expression was studied using real-time PCR. In PCa tissues, we found reduced levels of early tricarboxylic acid cycle metabolites, whereas the contents of urea cycle metabolites including aspartate, argininosuccinate, arginine, proline, and the oncometabolite fumarate were higher than that in benign controls. Fumarate content correlated positively with the gene expression of oncogenic HIF1α and NFκB pathways, which were significantly higher in the PCa samples than in the benign controls. Furthermore, data from the TCGA database demonstrated that prostate cancer patients with activated NFκB pathway had a lower survival rate. In summary, our data showed that fumarate content was positively associated with carcinogenic genes.
Collapse
Affiliation(s)
- Andras Franko
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (M.H.); (A.L.B.); (H.-U.H); (S.Z.L.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
| | - Yaping Shao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.S.); (C.H.); (X.L.); (X.Z.); (Q.W)
| | - Martin Heni
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (M.H.); (A.L.B.); (H.-U.H); (S.Z.L.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (T.T.); (A.S.)
| | - Miriam Hoene
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (M.H.); (A.P.)
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.S.); (C.H.); (X.L.); (X.Z.); (Q.W)
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.S.); (C.H.); (X.L.); (X.Z.); (Q.W)
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.S.); (C.H.); (X.L.); (X.Z.); (Q.W)
| | - Qingqing Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.S.); (C.H.); (X.L.); (X.Z.); (Q.W)
| | - Andreas L. Birkenfeld
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (M.H.); (A.L.B.); (H.-U.H); (S.Z.L.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
| | - Tilman Todenhöfer
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (T.T.); (A.S.)
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (T.T.); (A.S.)
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (M.H.); (A.P.)
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (M.H.); (A.L.B.); (H.-U.H); (S.Z.L.)
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
| | - Rainer Lehmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD), 72076 Tübingen, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (M.H.); (A.P.)
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.S.); (C.H.); (X.L.); (X.Z.); (Q.W)
| | - Stefan Z. Lutz
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (M.H.); (A.L.B.); (H.-U.H); (S.Z.L.)
- Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, 72116 Mössingen, Germany
| |
Collapse
|
22
|
Manguinhas R, Fernandes AS, Costa JG, Saraiva N, Camões SP, Gil N, Rosell R, Castro M, Miranda JP, Oliveira NG. Impact of the APE1 Redox Function Inhibitor E3330 in Non-small Cell Lung Cancer Cells Exposed to Cisplatin: Increased Cytotoxicity and Impairment of Cell Migration and Invasion. Antioxidants (Basel) 2020; 9:antiox9060550. [PMID: 32599967 PMCID: PMC7346157 DOI: 10.3390/antiox9060550] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 01/31/2023] Open
Abstract
Elevated expression levels of the apurinic/apyrimidinic endonuclease 1 (APE1) have been correlated with the more aggressive phenotypes and poor prognosis of non-small cell lung cancer (NSCLC). This study aimed to assess the impact of the inhibition of the redox function of APE1 with E3330 either alone or in combination with cisplatin in NSCLC cells. For this purpose, complementary endpoints focusing on cell viability, apoptosis, cell cycle distribution, and migration/invasion were studied. Cisplatin decreased the viability of H1975 cells in a time- and concentration-dependent manner, with IC50 values of 9.6 µM for crystal violet assay and 15.9 µM for 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay. E3330 was clearly cytotoxic for concentrations above 30 µM. The co-incubation of E3330 and cisplatin significantly decreased cell viability compared to cisplatin alone. Regarding cell cycle distribution, cisplatin led to an increase in sub-G1, whereas the co-treatment with E3330 did not change this profile, which was then confirmed in terms of % apoptotic cells. In addition, the combination of E3330 and cisplatin at low concentrations decreased collective and chemotactic migration, and also chemoinvasion, by reducing these capabilities up to 20%. Overall, these results point to E3330 as a promising compound to boost cisplatin therapy that warrants further investigation in NSCLC.
Collapse
Affiliation(s)
- Rita Manguinhas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (R.M.); (S.P.C.); (M.C.); (J.P.M.)
| | - Ana S. Fernandes
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal; (A.S.F.); (J.G.C.); (N.S.)
| | - João G. Costa
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal; (A.S.F.); (J.G.C.); (N.S.)
| | - Nuno Saraiva
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal; (A.S.F.); (J.G.C.); (N.S.)
| | - Sérgio P. Camões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (R.M.); (S.P.C.); (M.C.); (J.P.M.)
| | - Nuno Gil
- Lung Cancer Unit, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisboa, Portugal;
| | - Rafael Rosell
- Laboratory of Cellular and Molecular Biology, Institute for Health Science Research Germans Trias i Pujol (IGTP), Campus Can Ruti, Ctra de Can Ruti, Camí de les Escoles, s/n, 08916 Badalona, Barcelona, Spain;
- Internal Medicine Department, Universitat Autónoma de Barcelona, Campus de la UAB, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (R.M.); (S.P.C.); (M.C.); (J.P.M.)
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (R.M.); (S.P.C.); (M.C.); (J.P.M.)
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; (R.M.); (S.P.C.); (M.C.); (J.P.M.)
- Correspondence:
| |
Collapse
|
23
|
Hennigs JK, Minner S, Tennstedt P, Löser R, Huland H, Klose H, Graefen M, Schlomm T, Sauter G, Bokemeyer C, Honecker F. Subcellular Compartmentalization of Survivin is Associated with Biological Aggressiveness and Prognosis in Prostate Cancer. Sci Rep 2020; 10:3250. [PMID: 32094363 PMCID: PMC7039909 DOI: 10.1038/s41598-020-60064-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 01/29/2020] [Indexed: 11/09/2022] Open
Abstract
The role of subcellular survivin compartmentalization in the biology and prognosis of prostate cancer is unclear. We therefore investigated subcellular localization of survivin in more than 3000 prostate cancer patients by quantitative immunohistochemistry and performed transcriptomics of 250 prostate cancer patients and healthy donors using publicly available datasets. Survivin (BIRC5) gene expression was increased in primary prostate cancers and metastases, but did not differ in recurrent vs non-recurrent prostate cancers. Survivin immunohistochemistry (IHC) staining was limited exclusively to the nucleus in 900 prostate cancers (40.0%), and accompanied by various levels of cytoplasmic positivity in 1338 tumors (59.4%). 0.5% of prostate cancers did not express survivin. Nuclear and cytoplasmic survivin staining intensities were strongly associated with each other, pT category, and higher Gleason scores. Cytoplasmic but not nuclear survivin staining correlated with high tumor cell proliferation in prostate cancers. Strong cytoplasmic survivin staining, but not nuclear staining predicted an unfavorable outcome in univariate analyses. Multivariate Cox regression analysis showed that survivin is not an independent prognostic marker. In conclusion, we provide evidence that survivin expression is increased in prostate cancers, especially in metastatic disease, resulting in higher aggressiveness and tumor progression. In addition, subcellular compartmentalization is an important aspect of survivin cancer biology, as only cytoplasmic, but not nuclear survivin accumulation is linked to biological aggressiveness and prognosis of prostate cancers.
Collapse
Affiliation(s)
- Jan K Hennigs
- Department of Internal Medicine II - Oncology, Hematology, Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Institute of Pathology, University Medical Center Hamburg-Eppendorf, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pierre Tennstedt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Martini Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rolf Löser
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartwig Huland
- Martini Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans Klose
- Department of Internal Medicine II - Oncology, Hematology, Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Graefen
- Martini Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Martini Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Urology, Charité-Universitätsmedizin, Berlin, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Internal Medicine II - Oncology, Hematology, Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedemann Honecker
- Department of Internal Medicine II - Oncology, Hematology, Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Tumor and Breast Center ZeTuP, St. Gallen, Switzerland.
| |
Collapse
|
24
|
Codrich M, Comelli M, Malfatti MC, Mio C, Ayyildiz D, Zhang C, Kelley MR, Terrosu G, Pucillo CEM, Tell G. Inhibition of APE1-endonuclease activity affects cell metabolism in colon cancer cells via a p53-dependent pathway. DNA Repair (Amst) 2019; 82:102675. [PMID: 31450087 PMCID: PMC7092503 DOI: 10.1016/j.dnarep.2019.102675] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
The pathogenesis of colorectal cancer (CRC) involves different mechanisms, such as genomic and microsatellite instabilities. Recently, a contribution of the base excision repair (BER) pathway in CRC pathology has been emerged. In this context, the involvement of APE1 in the BER pathway and in the transcriptional regulation of genes implicated in tumor progression strongly correlates with chemoresistance in CRC and in more aggressive cancers. In addition, the APE1 interactome is emerging as an important player in tumor progression, as demonstrated by its interaction with Nucleophosmin (NPM1). For these reasons, APE1 is becoming a promising target in cancer therapy and a powerful prognostic and predictive factor in several cancer types. Thus, specific APE1 inhibitors have been developed targeting: i) the endonuclease activity; ii) the redox function and iii) the APE1-NPM1 interaction. Furthermore, mutated p53 is a common feature of advanced CRC. The relationship between APE1 inhibition and p53 is still completely unknown. Here, we demonstrated that the inhibition of the endonuclease activity of APE1 triggers p53-mediated effects on cell metabolism in HCT-116 colon cancer cell line. In particular, the inhibition of the endonuclease activity, but not of the redox function or of the interaction with NPM1, promotes p53 activation in parallel to sensitization of p53-expressing HCT-116 cell line to genotoxic treatment. Moreover, the endonuclease inhibitor affects mitochondrial activity in a p53-dependent manner. Finally, we demonstrated that 3D organoids derived from CRC patients are susceptible to APE1-endonuclease inhibition in a p53-status correlated manner, recapitulating data obtained with HCT-116 isogenic cell lines. These findings suggest the importance of further studies aimed at testing the possibility to target the endonuclease activity of APE1 in CRC.
Collapse
Affiliation(s)
- Marta Codrich
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Marina Comelli
- Laboratory of Bioenergetics, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Matilde Clarissa Malfatti
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Catia Mio
- Institute of Medical Genetics, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Dilara Ayyildiz
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Mark R Kelley
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and Pharmacology & Toxicology, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Giovanni Terrosu
- General Surgery and Transplantation Unit, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Carlo E M Pucillo
- Laboratory of Immunology, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy.
| |
Collapse
|
25
|
Fishel ML, Xia H, McGeown J, McIlwain DW, Elbanna M, Craft AA, Kaimakliotis HZ, Sandusky GE, Zhang C, Pili R, Kelley MR, Jerde TJ. Antitumor Activity and Mechanistic Characterization of APE1/Ref-1 Inhibitors in Bladder Cancer. Mol Cancer Ther 2019; 18:1947-1960. [PMID: 31413178 DOI: 10.1158/1535-7163.mct-18-1166] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/15/2019] [Accepted: 08/05/2019] [Indexed: 12/17/2022]
Abstract
Bladder cancer is the ninth most common cause of cancer-related deaths worldwide. Although cisplatin is used routinely in treating bladder cancer, refractory disease remains lethal for many patients. The recent addition of immunotherapy has improved patient outcomes; however, a large cohort of patients does not respond to these treatments. Therefore, identification of innovative molecular targets for bladder cancer is crucial. Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a multifunctional protein involved in both DNA repair and activation of transcription factors through reduction-oxidation (redox) regulation. High APE1/Ref-1 expression is associated with shorter patient survival time in many cancer types. In this study, we found high APE1/Ref-1 expression in human bladder cancer tissue relative to benign urothelium. Inhibition of APE1/Ref-1 redox signaling using APE1/Ref-1-specific inhibitors attenuates bladder cancer cell proliferation in monolayer, in three-dimensional cultures, and in vivo. This inhibition corresponds with an increase in apoptosis and decreased transcriptional activity of NF-κB and STAT3, transcription factors known to be regulated by APE1/Ref-1, resulting in decreased expression of downstream effectors survivin and Cyclin D1 in vitro and in vivo. We also demonstrate that in vitro treatment of bladder cancer cells with APE1/Ref-1 redox inhibitors in combination with standard-of-care chemotherapy cisplatin is more effective than cisplatin alone at inhibiting cell proliferation. Collectively, our data demonstrate that APE1/Ref-1 is a viable drug target for the treatment of bladder cancer, provide a mechanism of APE1/Ref-1 action in bladder cancer cells, and support the use of novel redox-selective APE1/Ref-1 inhibitors in clinical studies. SIGNIFICANCE: This work identifies a critical mechanism for APE1/Ref-1 in bladder cancer growth and provides compelling preclinical data using selective redox activity inhibitors of APE1/Ref-1 in vitro and in vivo.
Collapse
Affiliation(s)
- Melissa L Fishel
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hanyu Xia
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jack McGeown
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana.,University of Ulster, Coleraine, Northern Ireland
| | - David W McIlwain
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - May Elbanna
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ariel A Craft
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana
| | - Roberto Pili
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark R Kelley
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Travis J Jerde
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana. .,Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
26
|
APE1 stimulates EGFR-TKI resistance by activating Akt signaling through a redox-dependent mechanism in lung adenocarcinoma. Cell Death Dis 2018; 9:1111. [PMID: 30382076 PMCID: PMC6208429 DOI: 10.1038/s41419-018-1162-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/28/2018] [Accepted: 10/16/2018] [Indexed: 02/08/2023]
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have become the standard first-line treatment for advanced lung adenocarcinoma (LUAD) cancer patients with activating EGFR mutations. However, most patients show acquired resistance to EGFR-TKIs, thereby resulting in a modest overall survival benefit. Here, we found that expression level of APE1 was closely associated with TKI resistance in LUAD. Our clinical data show that level of APE1 was inversely correlated with progression-free survival rate and median time to progression in EGFR-mutated LUAD patients. Additionally, we observed increased expression of APE1 in TKI-resistant LUAD cell lines compared to their parental cell lines. Overexpression of APE1-protected TKI-sensitive LUAD cells from TKI-induced cell growth inhibition and cell death. In contrast, inhibition of APE1-enhanced TKI-induced apoptosis, cell growth inhibition and tumor growth inhibition in TKI-resistant LUAD. In addition, we identified that APE1 positively regulates Akt activation and APE1 overexpression-induced TKI resistance was attenuated by inhibition of Akt activity. Finally, we demonstrated that inhibition of the redox function of APE1 enhances the sensitivity of TKI-resistant LUAD cells to TKI treatment and inhibits Akt phosphorylation in TKI-resistant LUAD cells, but not by inhibition of the APE1 DNA repair function. Taken together, our data show that increased expression of APE1 significantly contributes to TKI resistance development in LUAD, and targeting APE1 may reverse acquired resistance of LUAD cells to TKI treatment. Additionally, our data show that APE1 regulates TKI resistance in LUAD cells by activating Akt signaling through a redox-dependent mechanism.
Collapse
|
27
|
Blocking HIF signaling via novel inhibitors of CA9 and APE1/Ref-1 dramatically affects pancreatic cancer cell survival. Sci Rep 2018; 8:13759. [PMID: 30214007 PMCID: PMC6137035 DOI: 10.1038/s41598-018-32034-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has reactive stroma that promotes tumor signaling, fibrosis, inflammation, and hypoxia, which activates HIF-1α to increase tumor cell metastasis and therapeutic resistance. Carbonic anhydrase IX (CA9) stabilizes intracellular pH following induction by HIF-1α. Redox effector factor-1 (APE1/Ref-1) is a multifunctional protein with redox signaling activity that converts certain oxidized transcription factors to a reduced state, enabling them to upregulate tumor-promoting genes. Our studies evaluate PDAC hypoxia responses and APE1/Ref-1 redox signaling contributions to HIF-1α-mediated CA9 transcription. Our previous studies implicated this pathway in PDAC cell survival under hypoxia. We expand those studies, comparing drug responses using patient-derived PDAC cells displaying differential hypoxic responses in 3D spheroid tumor-stroma models to characterize second generation APE1/Ref-1 redox signaling and CA9 inhibitors. Our data demonstrates that HIF-1α-mediated CA9 induction differs between patient-derived PDAC cells and that APE1/Ref-1 redox inhibition attenuates this induction by decreasing hypoxia-induced HIF-1 DNA binding. Dual-targeting of APE1/Ref-1 and CA9 in 3D spheroids demonstrated that this combination effectively kills PDAC tumor cells displaying drastically different levels of CA9. New APE1/Ref-1 and CA9 inhibitors were significantly more potent alone and in combination, highlighting the potential of combination therapy targeting the APE1-Ref-1 signaling axis with significant clinical potential.
Collapse
|
28
|
Sardar Pasha SPB, Sishtla K, Sulaiman RS, Park B, Shetty T, Shah F, Fishel ML, Wikel JH, Kelley MR, Corson TW. Ref-1/APE1 Inhibition with Novel Small Molecules Blocks Ocular Neovascularization. J Pharmacol Exp Ther 2018; 367:108-118. [PMID: 30076264 DOI: 10.1124/jpet.118.248088] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 08/01/2018] [Indexed: 01/11/2023] Open
Abstract
Ocular neovascular diseases like wet age-related macular degeneration are a major cause of blindness. Novel therapies are greatly needed for these diseases. One appealing antiangiogenic target is reduction-oxidation factor 1-apurinic/apyrimidinic endonuclease 1 (Ref-1/APE1). This protein can act as a redox-sensitive transcriptional activator for nuclear factor (NF)-κB and other proangiogenic transcription factors. An existing inhibitor of Ref-1's function, APX3330, previously showed antiangiogenic effects. Here, we developed improved APX3330 derivatives and assessed their antiangiogenic activity. We synthesized APX2009 and APX2014 and demonstrated enhanced inhibition of Ref-1 function in a DNA-binding assay compared with APX3330. Both compounds were antiproliferative against human retinal microvascular endothelial cells (HRECs; GI50 APX2009: 1.1 μM, APX2014: 110 nM) and macaque choroidal endothelial cells (Rf/6a; GI50 APX2009: 26 μM, APX2014: 5.0 μM). Both compounds significantly reduced the ability of HRECs and Rf/6a cells to form tubes at mid-nanomolar concentrations compared with control, and both significantly inhibited HREC and Rf/6a cell migration in a scratch wound assay, reducing NF-κB activation and downstream targets. Ex vivo, APX2009 and APX2014 inhibited choroidal sprouting at low micromolar and high nanomolar concentrations, respectively. In the laser-induced choroidal neovascularization mouse model, intraperitoneal APX2009 treatment significantly decreased lesion volume by 4-fold compared with vehicle (P < 0.0001, ANOVA with Dunnett's post-hoc tests), without obvious intraocular or systemic toxicity. Thus, Ref-1 inhibition with APX2009 and APX2014 blocks ocular angiogenesis in vitro and ex vivo, and APX2009 is an effective systemic therapy for choroidal neovascularization in vivo, establishing Ref-1 inhibition as a promising therapeutic approach for ocular neovascularization.
Collapse
Affiliation(s)
- Sheik Pran Babu Sardar Pasha
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Kamakshi Sishtla
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Bomina Park
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Trupti Shetty
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Fenil Shah
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Melissa L Fishel
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - James H Wikel
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Mark R Kelley
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology (S.P.B.S.P., K.S., R.S.S., B.P., T.S., T.W.C.), Department of Pharmacology and Toxicology (R.S.S., B.P., T.S., M.L.F., M.R.K., T.W.C.), Department of Biochemistry and Molecular Biology (M.R.K., T.W.C.), Herman B Wells Center for Pediatric Research, Department of Pediatrics (F.S., M.L.F., M.R.K.), and Melvin and Bren Simon Cancer Center (M.L.F., M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana; and Apexian Pharmaceuticals (J.H.W.), Indianapolis, Indiana
| |
Collapse
|
29
|
Abstract
Reduction-oxidation factor 1-apurinic/apyrimidinic endonuclease (Ref-1/APE1) is a critical node in tumor cells, both as a redox regulator of transcription factor activation and as part of the DNA damage response. As a redox signaling protein, Ref-1/APE1 enhances the transcriptional activity of STAT3, HIF-1α, nuclear factor kappa B, and other transcription factors to promote growth, migration, and survival in tumor cells as well as inflammation and angiogenesis in the tumor microenvironment. Ref-1/APE1 is activated in a variety of cancers, including prostate, colon, pancreatic, ovarian, lung and leukemias, leading to increased aggressiveness. Transcription factors downstream of Ref-1/APE1 are key contributors to many cancers, and Ref-1/APE1 redox signaling inhibition slows growth and progression in a number of tumor types. Ref-1/APE1 inhibition is also highly effective when paired with other drugs, including standard-of-care therapies and therapies targeting pathways affected by Ref-1/APE1 redox signaling. Additionally, Ref-1/APE1 plays a role in a variety of other indications, such as retinopathy, inflammation, and neuropathy. In this review, we discuss the functional consequences of activation of the Ref-1/APE1 node in cancer and other diseases, as well as potential therapies targeting Ref-1/APE1 and related pathways in relevant diseases. APX3330, a novel oral anticancer agent and the first drug to target Ref-1/APE1 for cancer is entering clinical trials and will be explored in various cancers and other diseases bringing bench discoveries to the clinic.
Collapse
|