1
|
Yu Y, Cen C, Shao Z, Wang C, Wang Y, Miao Z, Sun M, Wang C, Xu Q, Liang K, Zhou J, Zhou D, Ji H, Xu G, Du Y. APE1 promotes lung adenocarcinoma through G4-mediated transcriptional reprogramming of urea cycle metabolism. iScience 2025; 28:112275. [PMID: 40276763 PMCID: PMC12019196 DOI: 10.1016/j.isci.2025.112275] [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: 01/24/2024] [Revised: 07/17/2024] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
Lung adenocarcinoma (LUAD) remains the leading cause of cancer deaths worldwide. Apurinic/apyrimidinic endonuclease 1 (APE1), an enzyme integral to DNA repair and redox signaling, is notably upregulated in LUAD. Here we reveal that APE1 amplification, primarily via allele duplication, strongly correlates with poor prognosis in LUAD patients. Using human LUAD cell lines and a KRAS-driven mouse model, we showed that APE1 deletion hampered cell proliferation and tumor growth, highlighting its role in tumorigenesis. Mechanistically, APE1 promoted the transcription of urea cycle genes CPS1 and ARG2 by modulating the presence of G-quadruplex (G4) structures in their promoter regions. APE1 loss disrupted the urea cycle and pyrimidine metabolism, inducing metabolic reprogramming and growth arrest, which could be rescued by CPS1 or pyrimidine restoration. These findings uncover APE1's role in transcriptional regulation of urea cycle metabolic reprogramming via G4 structure, providing a potential therapeutic target LUAD patients with elevated APE1 expression.
Collapse
Affiliation(s)
- Yanhao Yu
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chaochao Cen
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenyu Shao
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chaohan Wang
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yiqin Wang
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zongjie Miao
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Meiling Sun
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Wang
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing Xu
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwei Liang
- School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Jiaxin Zhou
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Zhou
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Chinese Academy of Medical Sciences (RU069) & Zhongshan-Xuhui Hospital, Medical College of Fudan University, Shanghai 200032, China
| | - Hongbin Ji
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoliang Xu
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Chinese Academy of Medical Sciences (RU069) & Zhongshan-Xuhui Hospital, Medical College of Fudan University, Shanghai 200032, China
| | - Yarui Du
- CAS Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
2
|
Kelley MR, Wan J, Liu S, Kpenu E, Wireman R, Mosley AL, Liu H, Lakhani NJ, Shahda S, O'Neil B, Opyrchal M, Messmann RA. A Phase I study targeting the APE1/Ref-1 redox signaling protein with APX3330: First clinical agent targeting APE1/Ref-1 in Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.03.25325173. [PMID: 40236444 PMCID: PMC11998840 DOI: 10.1101/2025.04.03.25325173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Purpose APX3330 is an oral agent targeting the redox signaling activity of Ape1/Ref-1 (Ref-1), a key regulator of transcription factors involved in inflammation and tumorigenesis. APX3330 selectively inhibits Ref-1's redox function without affecting its DNA repair role. This Phase 1, multicenter, open-label, dose-escalation study in advanced solid tumor was aimed at determining the recommended Phase 2 dose (RP2D) while assessing safety, pharmacokinetics, and biomarker evidence of target engagement. Clinical trial: NCT03375086 . Patients and Methods Nineteen cancer patients were treated, with eight completing follow-up. Subjects received APX3330 orally twice daily in 21-day cycles, starting at 240 mg/day and escalating in 120 mg/day increments. Adverse event (AE) monitoring followed a 1 pt/cohort approach until a >G2 toxicity event, after which a 3+3 design was implemented. Treatment continued until disease progression, consent withdrawal, or intolerable toxicity. Antitumor activity was assessed using RECIST 1.1, and pharmacodynamic markers included serum Ref-1 levels and circulating tumor cells. Results Six subjects had stable disease for >4 cycles, with four remaining on study for 252- 421 days. No treatment-related serious adverse events occurred. One subject (720 mg cohort) withdrew due to Grade 3 maculopapular rash (dose-limiting toxicity). Laboratory assessments and ECGs showed no clinically significant abnormalities. Conclusions APX3330 demonstrated clinical benefit by stabilizing disease in ∼33% of subjects. Ref-1 target engagement was confirmed via biomarker analyses, with reduced serum Ref-1 and circulating tumor cells. The RP2D is 600 mg daily, with APX3330 showing a favorable safety profile and target-mediated effects.
Collapse
|
3
|
Yuan HH, Yin H, Marincas M, Xie LL, Bu LL, Guo MH, Zheng XL. From DNA Repair to Redox Signaling: The Multifaceted Role of APEX1 (Apurinic/Apyrimidinic Endonuclease 1) in Cardiovascular Health and Disease. Int J Mol Sci 2025; 26:3034. [PMID: 40243693 PMCID: PMC11988304 DOI: 10.3390/ijms26073034] [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: 02/13/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Apurinic/apyrimidinic endonuclease 1 (APEX1) serves as a potent regulatory factor in innate immunity, exhibiting both redox and endonuclease activities. Its redox function enables the regulation of transcription factors such as NF-κB or STAT3, whereas its endonuclease activity recognizes apurinic/apyrimidinic (AP) sites in damaged DNA lesions during base excision repair (BER) and double-stranded DNA repair, thereby I confirm.anti-inflammatory, antioxidative stress and antiapoptotic effects. APEX1 is expressed in a variety of cell types that constitute the cardiovascular system, including cardiomyocytes, endothelial cells, smooth muscle cells, and immune cells. Emerging genetic and experimental evidence points towards the functional roles of APEX1 in the pathophysiology of cardiovascular diseases, including neointimal formation and atherosclerosis. This review aims to present comprehensive coverage of the up-to-date literature concerning the molecular and cellular functions of APEX1, with a particular focus on how APEX1 contributes to the (dys)functions of different cell types during the pathogenesis of cardiovascular diseases. Furthermore, we underscore the potential of APEX1 as a therapeutic target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Huan-Huan Yuan
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond St. N., London, ON N6A 5B7, Canada
| | - Mara Marincas
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Ling-Li Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Lan-Lan Bu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Min-Hua Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xi-Long Zheng
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| |
Collapse
|
4
|
Hartman GD, Muniyandi A, Sishtla K, Kpenu EK, Miller WP, Kaplan BA, Kim LA, Liu S, Wan J, Qi X, Boulton ME, Kelley MR, Corson TW. Ref-1 redox activity regulates retinal neovascularization by modulating transcriptional activation of HIF-1α. FASEB J 2025; 39:e70348. [PMID: 39902892 PMCID: PMC11792779 DOI: 10.1096/fj.202401989rr] [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: 08/29/2024] [Revised: 12/12/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Retinal neovascularization impairs visual function and is a hallmark of several neovascular eye diseases, including retinopathy of prematurity (ROP) and proliferative diabetic retinopathy (PDR). Current treatments include intravitreal injections of anti-vascular endothelial growth factor (VEGF) biologics, but these therapeutics are often accompanied by high treatment burden and resistance to therapy. Prior studies indicate that APE1/Ref-1, a multifunctional protein with both endonuclease (APE1) and redox-mediated transcriptional regulatory activity (Ref-1), activates multiple pro-angiogenic and pro-inflammatory signaling pathways by chemically reducing key cysteine residues in transcription factors, thereby activating them. Here, we investigated the previously unexplored role of Ref-1 in retinal neovascularization. We demonstrate that Ref-1 is highly expressed in endothelial cells in human PDR and in the oxygen-induced retinopathy (OIR) mouse model of retinal neovascularization. Ref-1 is also highly expressed in microglia and astrocytes in OIR. A small molecule Ref-1 redox inhibitor, APX2009, decreased retinal neovascularization in OIR after systemic delivery. In vitro, hypoxic endothelial cells did not exhibit upregulation of Ref-1 but rather increased Ref-1 nuclear localization. APX2009 decreased hypoxic endothelial cell proliferation and HIF-1α transcriptional activation. Thus, Ref-1 redox activity may be a novel therapeutic target for the treatment of retinal neovascularization, making APX2009 a promising systemic therapeutic approach for the treatment of vascular retinopathies such as ROP and PDR.
Collapse
Affiliation(s)
- Gabriella D. Hartman
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Anbukkarasi Muniyandi
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kamakshi Sishtla
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisIndianaUSA
- Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
| | - Eyram K. Kpenu
- Herman B Wells Center for Pediatric Research, Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisIndianaUSA
| | - William P. Miller
- Schepens Eye Research Institute of Mass Eye and EarHarvard Medical SchoolBostonMassachusettsUSA
| | - Bryan A. Kaplan
- Schepens Eye Research Institute of Mass Eye and EarHarvard Medical SchoolBostonMassachusettsUSA
| | - Leo A. Kim
- Schepens Eye Research Institute of Mass Eye and EarHarvard Medical SchoolBostonMassachusettsUSA
| | - Sheng Liu
- Center for Computational Biology and BioinformaticsIndiana UniversityIndianapolisIndianaUSA
| | - Jun Wan
- Center for Computational Biology and BioinformaticsIndiana UniversityIndianapolisIndianaUSA
| | - Xiaoping Qi
- Department of Ophthalmology and Visual SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Michael E. Boulton
- Department of Ophthalmology and Visual SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Mark R. Kelley
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisIndianaUSA
- Herman B Wells Center for Pediatric Research, Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisIndianaUSA
| | - Timothy W. Corson
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisIndianaUSA
- Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
- Indiana University Simon Comprehensive Cancer CenterIndianapolisIndianaUSA
- Department of Ophthalmology and Vision Sciences, Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
5
|
Muniyandi A, Hartman GD, Sishtla K, Rai R, Gomes C, Day K, Song Y, Masters AR, Quinney SK, Qi X, Woods H, Boulton ME, Meyer JS, Vilseck JZ, Georgiadis MM, Kelley MR, Corson TW. Ref-1 is overexpressed in neovascular eye disease and targetable with a novel inhibitor. Angiogenesis 2025; 28:11. [PMID: 39756006 PMCID: PMC12019292 DOI: 10.1007/s10456-024-09966-0] [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: 07/09/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
Reduction-oxidation factor-1 or apurinic/apyrimidinic endonuclease 1 (Ref-1/APE1) is a crucial redox-sensitive activator of transcription factors such as NF-κB, HIF-1α, STAT-3 and others. It could contribute to key features of ocular neovascularization including inflammation and angiogenesis; these underlie diseases like neovascular age-related macular degeneration (nAMD). We previously revealed a role for Ref-1 in the growth of ocular endothelial cells and in choroidal neovascularization (CNV). Here, we set out to further explore Ref-1 in neovascular eye disease. Ref-1 was highly expressed in human nAMD, murine laser-induced CNV and Vldlr-/- mouse subretinal neovascularization (SRN). Ref-1's interaction with a redox-specific small molecule inhibitor, APX2009, was shown by NMR and docking. This compound blocks crucial angiogenic features in multiple endothelial cell types. APX2009 also ameliorated murine laser-induced choroidal neovascularization (L-CNV) when delivered intravitreally. Moreover, systemic APX2009 reduced murine SRN and downregulated the expression of Ref-1 redox regulated HIF-1α target carbonic anhydrase 9 (CA9) in the Vldlr-/- mouse model. Our data validate the redox function of Ref-1 as a critical regulator of ocular angiogenesis, indicating that inhibition of Ref-1 holds therapeutic potential for treating nAMD.
Collapse
Affiliation(s)
- Anbukkarasi Muniyandi
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gabriella D Hartman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Kamakshi Sishtla
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Ratan Rai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kristina Day
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yang Song
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andi R Masters
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara K Quinney
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Xiaoping Qi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hailey Woods
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason S Meyer
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonah Z Vilseck
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Millie M Georgiadis
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Mark R Kelley
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Timothy W Corson
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
- Department of Ophthalmology and Vision Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Hu M, Zhang Y, Zhang P, Liu K, Zhang M, Li L, Yu Z, Zhang X, Zhang W, Xu Y. Targeting APE1: Advancements in the Diagnosis and Treatment of Tumors. Protein Pept Lett 2025; 32:18-33. [PMID: 39648425 DOI: 10.2174/0109298665338519241114103223] [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: 07/11/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 12/10/2024]
Abstract
With the emergence of the precision medicine era, targeting specific proteins has emerged as a pivotal breakthrough in tumor diagnosis and treatment. Apurinic/apyrimidinic Endonuclease 1 (APE1) is a multifunctional protein that plays a crucial role in DNA repair and cellular redox regulation. This article comprehensively explores the fundamental mechanisms of APE1 as a multifunctional enzyme in biology, with particular emphasis on its potential significance in disease diagnosis and strategies for tumor treatment. Firstly, this article meticulously analyzes the intricate biological functions of APE1 at a molecular level, establishing a solid theoretical foundation for subsequent research endeavors. In terms of diagnostic applications, the presence of APE1 can be detected in patient serum samples, biopsy tissues, and through cellular in situ testing. The precise detection methods enable changes in APE1 levels to serve as reliable biomarkers for predicting tumor occurrence, progression, and patient prognosis. Moreover, this article focuses on elucidating the potential role of APE1 in tumor treatment by exploring various inhibitors, including nucleic acid-based inhibitors and small molecule drug inhibitors categories, and revealing their unique advantages in disrupting DNA repair function and modulating oxidative-reduction activity. Finally, the article provides an outlook on future research directions for APE1 while acknowledging major technical difficulties and clinical challenges that need to be overcome despite its immense potential as a target for tumor therapy.
Collapse
Affiliation(s)
- Minghui Hu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Yingyu Zhang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Kangbo Liu
- Henan Institute for Drug and Medical Device Inspection (Henan Vaccine Issuance Center), Zhengzhou, 450018, China
| | - Mengxin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Lifeng Li
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Zhidan Yu
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Xianwei Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Wancun Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Ying Xu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| |
Collapse
|
7
|
Lalunio H, Stupka N, Goodman CA, Hayes A. The Potential of Targeting APE1/Ref-1 as a Therapeutic Intervention for Duchenne Muscular Dystrophy. Antioxid Redox Signal 2024. [PMID: 39729027 DOI: 10.1089/ars.2024.0620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Significance: Inflammation and oxidative stress play crucial roles in the development and progression of skeletal muscle diseases. This review aims to examine the existing evidence regarding the involvement and inhibition of APE1/Ref-1 (apurinic/apyrimidinic endonuclease 1/redox factor 1) in diseases, then extrapolate this evidence to the context of skeletal muscle and discuss the potential beneficial effects of APE1/Ref-1 inhibition in ameliorating myopathy with a particular focus on dystrophic pathology. Critical Issues: Currently, therapeutic interventions targeting pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and nuclear factor erythroid 2-related factor 2 (NRF2), have shown limited efficacy in both clinical and preclinical settings. Thus, there is a need for a more comprehensive treatment approach. Recent Advances: APE1/Ref-1 is a multifunctional protein that was initially identified as being involved in DNA repair. However, newer research has revealed its additional role as a redox-sensitive regulator of transcription factors, including NF-κB and NRF2. Numerous studies have reported increased expression of APE1/Ref-1 in various disorders and have demonstrated the beneficial effects of inhibiting its redox function using the small molecular inhibitor, APX3330. Although these pathways are similarly dysregulated in neuromuscular disorders, the specific role of APE1/Ref-1 in skeletal muscle remains unclear, with only a limited number of studies noting its presence in this tissue. Future Directions: Further studies investigating the role of APE1/Ref-1 in skeletal muscle and identifying whether APE1/Ref-1 is up- or downregulated in dystrophic skeletal muscle would be required to determine whether upregulating or inhibiting the redox function of APE1/Ref-1 will alleviate chronic inflammation and heightened oxidative stress. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Hannah Lalunio
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
| | - Nicole Stupka
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
| | - Craig A Goodman
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
- Institute of Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Centre for Muscle Research, The University of Melbourne, Parkville, Australia
| | - Alan Hayes
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
- Institute of Health and Sport (IHeS), Victoria University, Melbourne, Australia
| |
Collapse
|
8
|
Zhao H, Richardson C, Marriott I, Yang IH, Yan S. APE1 is a master regulator of the ATR-/ATM-mediated DNA damage response. DNA Repair (Amst) 2024; 144:103776. [PMID: 39461278 PMCID: PMC11611674 DOI: 10.1016/j.dnarep.2024.103776] [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: 09/18/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
To maintain genomic integrity, cells have evolved several conserved DNA damage response (DDR) pathways in response to DNA damage and stress conditions. Apurinic/apyrimidinic endonuclease 1 (APE1) exhibits AP endonuclease, 3'-5' exonuclease, 3'-phosphodiesterase, and 3'-exoribonuclease activities and plays critical roles in the DNA repair and redox regulation of transcription. However, it remains unclear whether and how APE1 is involved in DDR pathways. In this perspective, we first updated our knowledge of APE1's functional domains and its nuclease activities and their specific associated substrates. We then summarized the newly discovered roles and mechanisms of action of APE1 in the global and nucleolar ATR-mediated DDR pathway. While the ATM-mediated DDR is well known to be activated by DNA double-strand breaks and oxidative stress, here we provided new perspectives as to how ATM DDR signaling is activated by indirect single-strand breaks (SSBs) resulting from genotoxic stress and defined SSB structures, and discuss how ATM kinase is directly activated and regulated by its activator, APE1. Together, accumulating body of new evidence supports the notion that APE1 is a master regulator protein of the ATR- and ATM-mediated DDR pathways. These new findings of APE1 in DDR signaling provide previously uncharacterized but critical functions and regulations of APE1 in genome integrity.
Collapse
Affiliation(s)
- Haichao Zhao
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Christine Richardson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; School of Data Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - In Hong Yang
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Shan Yan
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; School of Data Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
9
|
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
|
10
|
Meng F, Li T, Singh AK, Wang Y, Attiyeh M, Kohram F, Feng Q, Li YR, Shen B, Williams T, Liu Y, Raoof M. Base-excision repair pathway regulates transcription-replication conflicts in pancreatic ductal adenocarcinoma. Cell Rep 2024; 43:114820. [PMID: 39368091 DOI: 10.1016/j.celrep.2024.114820] [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: 11/24/2023] [Revised: 07/19/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024] Open
Abstract
Oncogenic mutations (such as in KRAS) can dysregulate transcription and replication, leading to transcription-replication conflicts (TRCs). Here, we demonstrate that TRCs are enriched in human pancreatic ductal adenocarcinoma (PDAC) compared to other common solid tumors or normal cells. Several orthogonal approaches demonstrated that TRCs are oncogene dependent. A small interfering RNA (siRNA) screen identified several factors in the base-excision repair (BER) pathway as main regulators of TRCs in PDAC cells. Inhibitors of BER pathway (methoxyamine and CRT) enhanced TRCs. Mechanistically, BER pathway inhibition severely altered RNA polymerase II (RNAPII) and R-loop dynamics at nascent DNA, causing RNAPII trapping and contributing to enhanced TRCs. The ensuing DNA damage activated the ATR-Chk1 pathway. Co-treatment with ATR inhibitor (VX970) and BER inhibitor (methoxyamine) at clinically relevant doses synergistically enhanced DNA damage and reduced cell proliferation in PDAC cells. The study provides mechanistic insights into the regulation of TRCs in PDAC by the BER pathway, which has biologic and therapeutic implications.
Collapse
Affiliation(s)
- Fan Meng
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Tiane Li
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA, USA; Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Yingying Wang
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Marc Attiyeh
- Department of Surgery, Cedars Sinai, Los Angeles, CA, USA
| | - Fatemeh Kohram
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Qianhua Feng
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yun R Li
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Binghui Shen
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Terence Williams
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yilun Liu
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA; Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
11
|
Xiao H, Jiang N, Zhang H, Wang S, Pi Q, Chen H, He X, Luo W, Lu Y, Deng Y, Zhong Z. Inhibitors of APE1 redox and ATM synergistically sensitize osteosarcoma cells to ionizing radiation by inducing ferroptosis. Int Immunopharmacol 2024; 139:112672. [PMID: 39032469 DOI: 10.1016/j.intimp.2024.112672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
The resistance of osteosarcoma (OS) to ionizing radiation (IR) is an obstacle for effective patient treatment. Apurinic/apyrimidinic endonuclease-reduction/oxidation factor 1 (APE1/Ref-1) is a multifunctional protein with DNA repair and reduction/oxidation (redox) activities. We previously revealed the role of APE1 in OS radioresistance; however, whether the redox activity of APE1 is involved in OS radioresistance is unclear. APE1 regulates the activation of ataxia-telangiectasia mutated (ATM), an initiator of DNA damage response that mediates radioresistance in other cancers. The role of APE1 redox activity and ATM activation in OS radioresistance is unknown. Our study revealed that IR increased APE1 expression and ATM activation in OS cells, and APE1 directly regulated ATM activation by its redox activity. The combined use of an APE1 redox inhibitor and ATM inhibitor effectively sensitized OS cells to IR in vitro and in vivo. Mechanistically, the increased radiosensitization of OS cells by the combined use of the two inhibitors was mediated by increased ferroptosis. Co-treatment with the two inhibitors significantly decreased expression of the common targeted transcription factor P53 compared with single inhibitor treatment. Collectively, APE1 redox activity, ATM activation and their crosstalk play important roles in the resistance of OS to irradiation. Synergetic inhibition of APE1 redox activity and ATM activation sensitized OS cells to IR by inducing ferroptosis, which provides a promising strategy for OS radiotherapy.
Collapse
Affiliation(s)
- Hanxi Xiao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, China; Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, China; Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Nan Jiang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Hongbin Zhang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Shuai Wang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qin Pi
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Huawei Chen
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xuan He
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei Luo
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yonghui Lu
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing 400038, China.
| | - Youcai Deng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, China; Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, China.
| | - Zhaoyang Zhong
- Cancer Center, Daping Hospital, Army Medical University, Chongqing 400042, China; Department of Oncology, The Fifth People's Hospital of Chongqing, Chongqing 400062, China.
| |
Collapse
|
12
|
Liu Z, Yan B, Liu H, Liu X, Xiao X, Ming Z. Enhancing APE1 detection through apurinic/apyrimidinic site inhibition of DNA polymerase: an innovative, highly sensitive approach. Chem Commun (Camb) 2024; 60:4695-4698. [PMID: 38592754 DOI: 10.1039/d4cc00304g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
This study presents an innovative method for the highly sensitive detection of apurinic/apyrimidinic endonuclease 1 (APE1), a crucial biomarker and target for cancer diagnosis and treatment. The method is predicated on our discovery that the apurinic or apyrimidinic site (AP site) can inhibit the activity of Taq DNA polymerase. Subsequent experiments further led to the development of a new amplification method based on the digestion activity of Lambda exonuclease. This approach showed potential to detect trace amounts of APE1 in biological samples with high sensitivity.
Collapse
Affiliation(s)
- Zhijun Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Bei Yan
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Huan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiao Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xianjin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhihao Ming
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China.
| |
Collapse
|
13
|
Mao S, Xie C, Liu Y, Zhao Y, Li M, Gao H, Xiao Y, Zou Y, Zheng Z, Gao Y, Xie J, Tian B, Wang L, Hua Y, Xu H. Apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) promotes stress granule formation via YBX1 phosphorylation in ovarian cancer. Cell Mol Life Sci 2024; 81:113. [PMID: 38436697 PMCID: PMC10912283 DOI: 10.1007/s00018-023-05086-y] [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: 09/08/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 03/05/2024]
Abstract
APE1 is an essential gene involved in DNA damage repair, the redox regulation of transcriptional factors (TFs) and RNA processing. APE1 overexpression is common in cancers and correlates with poor patient survival. Stress granules (SGs) are phase-separated cytoplasmic assemblies that cells form in response to environmental stresses. Precise regulation of SGs is pivotal to cell survival, whereas their dysregulation is increasingly linked to diseases. Whether APE1 engages in modulating SG dynamics is worthy of investigation. In this study, we demonstrate that APE1 colocalizes with SGs and promotes their formation. Through phosphoproteome profiling, we discover that APE1 significantly alters the phosphorylation landscape of ovarian cancer cells, particularly the phosphoprofile of SG proteins. Notably, APE1 promotes the phosphorylation of Y-Box binding protein 1 (YBX1) at S174 and S176, leading to enhanced SG formation and cell survival. Moreover, expression of the phosphomutant YBX1 S174/176E mimicking hyperphosphorylation in APE1-knockdown cells recovered the impaired SG formation. These findings shed light on the functional importance of APE1 in SG regulation and highlight the importance of YBX1 phosphorylation in SG dynamics.
Collapse
Affiliation(s)
- Shuyu Mao
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Chong Xie
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
- Department of Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yufeng Liu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Ye Zhao
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Mengxia Li
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinses Academy of Sciences, Hangzhou, China
| | - Han Gao
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinses Academy of Sciences, Hangzhou, China
| | - Yue Xiao
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Yongkang Zou
- Department of Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhiguo Zheng
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Ya Gao
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Juan Xie
- Department of Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Bing Tian
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Liangyan Wang
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China
| | - Yuejin Hua
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China.
| | - Hong Xu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, Institute of Biophysics, College of Life Science, Zhejiang University, Hangzhou, China.
| |
Collapse
|
14
|
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
|
15
|
Kpenu EK, Kelley MR. Combating PDAC Drug Resistance: The Role of Ref-1 Inhibitors in Accelerating Progress in Pancreatic Cancer Research. JOURNAL OF CELLULAR SIGNALING 2024; 5:208-216. [PMID: 39635662 PMCID: PMC11616473 DOI: 10.33696/signaling.5.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most lethal solid tumor diagnoses given its limited treatment options and dismal prognosis. Its complex tumor microenvironment (TME), heterogeneity, and high propensity for drug resistance are major obstacles in developing effective therapies. Here, we highlight the critical role of Redox effector 1 (Ref-1) in PDAC progression and drug resistance, focusing on its redox regulation of key transcription factors (TFs) such as STAT3, HIF1α, and NF-κB, which are pivotal for tumor survival, proliferation, and immune evasion. We discuss the development of novel Ref-1 inhibitors, including second-generation compounds with enhanced potency and improved pharmacokinetic profiles, which have shown significant promise in preclinical models. These inhibitors disrupt Ref-1's redox function, leading to decreased TF activity and increased chemosensitivity in PDAC cells. We further detail our utilization of advanced preclinical models, such as 3D spheroids, organoids, and Tumor-Microenvironment-on-Chip (T-MOC) systems, which better simulate the complex conditions of the PDAC TME and improve the predictive power of therapeutic responses. By targeting Ref-1 and its associated pathways, in conjunction with improved models, more replicative of PDAC's TME, we are focused on approaches which hold the potential to overcome current therapeutic limitations and advance the development of more effective treatments for PDAC. Our findings suggest that integrating Ref-1 inhibitors into combination therapies could disrupt multiple survival mechanisms within the tumor, offering new hope for improving outcomes in this challenging cancer.
Collapse
Affiliation(s)
- Eyram K. Kpenu
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University School of Medicine, Department of Biochemistry and Molecular Biology, Indianapolis, IN, USA
| | - Mark 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
| |
Collapse
|
16
|
Malfatti MC, Bellina A, Antoniali G, Tell G. Revisiting Two Decades of Research Focused on Targeting APE1 for Cancer Therapy: The Pros and Cons. Cells 2023; 12:1895. [PMID: 37508559 PMCID: PMC10378182 DOI: 10.3390/cells12141895] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
APE1 is an essential endodeoxyribonuclease of the base excision repair pathway that maintains genome stability. It was identified as a pivotal factor favoring tumor progression and chemoresistance through the control of gene expression by a redox-based mechanism. APE1 is overexpressed and serum-secreted in different cancers, representing a prognostic and predictive factor and a promising non-invasive biomarker. Strategies directly targeting APE1 functions led to the identification of inhibitors showing potential therapeutic value, some of which are currently in clinical trials. Interestingly, evidence indicates novel roles of APE1 in RNA metabolism that are still not fully understood, including its activity in processing damaged RNA in chemoresistant phenotypes, regulating onco-miRNA maturation, and oxidized RNA decay. Recent data point out a control role for APE1 in the expression and sorting of onco-miRNAs within secreted extracellular vesicles. This review is focused on giving a portrait of the pros and cons of the last two decades of research aiming at the identification of inhibitors of the redox or DNA-repair functions of APE1 for the definition of novel targeted therapies for cancer. We will discuss the new perspectives in cancer therapy emerging from the unexpected finding of the APE1 role in miRNA processing for personalized therapy.
Collapse
Affiliation(s)
- Matilde Clarissa Malfatti
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Alessia Bellina
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Giulia Antoniali
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
17
|
Muniyandi A, Hartman GD, Song Y, Mijit M, Kelley MR, Corson TW. Beyond VEGF: Targeting Inflammation and Other Pathways for Treatment of Retinal Disease. J Pharmacol Exp Ther 2023; 386:15-25. [PMID: 37142441 PMCID: PMC10289243 DOI: 10.1124/jpet.122.001563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Neovascular eye diseases include conditions such as retinopathy of prematurity, proliferative diabetic retinopathy, and neovascular age-related macular degeneration. Together, they are a major cause of vision loss and blindness worldwide. The current therapeutic mainstay for these diseases is intravitreal injections of biologics targeting vascular endothelial growth factor (VEGF) signaling. Lack of universal response to these anti-VEGF agents coupled with the challenging delivery method underscore a need for new therapeutic targets and agents. In particular, proteins that mediate both inflammatory and proangiogenic signaling are appealing targets for new therapeutic development. Here, we review agents currently in clinical trials and highlight some promising targets in preclinical and early clinical development, focusing on the redox-regulatory transcriptional activator APE1/Ref-1, the bioactive lipid modulator soluble epoxide hydrolase, the transcription factor RUNX1, and others. Small molecules targeting each of these proteins show promise for blocking neovascularization and inflammation. The affected signaling pathways illustrate the potential of new antiangiogenic strategies for posterior ocular disease. SIGNIFICANCE STATEMENT: Discovery and therapeutic targeting of new angiogenesis mediators is necessary to improve treatment of blinding eye diseases like retinopathy of prematurity, diabetic retinopathy, and neovascular age-related macular degeneration. Novel targets undergoing evaluation and drug discovery work include proteins important for both angiogenesis and inflammation signaling, including APE1/Ref-1, soluble epoxide hydrolase, RUNX1, and others.
Collapse
Affiliation(s)
- Anbukkarasi Muniyandi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Gabriella D Hartman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Yang Song
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Mahmut Mijit
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark R Kelley
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Timothy W Corson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
18
|
Mijit M, Liu S, Sishtla K, Hartman GD, Wan J, Corson TW, Kelley MR. Identification of Novel Pathways Regulated by APE1/Ref-1 in Human Retinal Endothelial Cells. Int J Mol Sci 2023; 24:1101. [PMID: 36674619 PMCID: PMC9865623 DOI: 10.3390/ijms24021101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/26/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
APE1/Ref-1 (apurinic/apyrimidinic endonuclease 1, APE1 or APEX1; redox factor-1, Ref-1) is a dual-functional enzyme with crucial roles in DNA repair, reduction/oxidation (redox) signaling, and RNA processing and metabolism. The redox function of Ref-1 regulates several transcription factors, such as NF-κB, STAT3, HIF-1α, and others, which have been implicated in multiple human diseases, including ocular angiogenesis, inflammation, and multiple cancers. To better understand how APE1 influences these disease processes, we investigated the effects of APEX1 knockdown (KD) on gene expression in human retinal endothelial cells. This abolishes both DNA repair and redox signaling functions, as well as RNA interactions. Using RNA-seq analysis, we identified the crucial signaling pathways affected following APEX1 KD, with subsequent validation by qRT-PCR. Gene expression data revealed that multiple genes involved in DNA base excision repair, other DNA repair pathways, purine or pyrimidine metabolism signaling, and histidine/one carbon metabolism pathways were downregulated by APEX1 KD. This is in contrast with the alteration of pathways by APEX1 KD in human cancer lines, such as pancreatic ductal adenocarcinoma, lung, HeLa, and malignant peripheral nerve sheath tumors. These results highlight the unique role of APE1/Ref-1 and the clinical therapeutic potential of targeting APE1 and pathways regulated by APE1 in the eye. These findings provide novel avenues for ocular neovascularization treatment.
Collapse
Affiliation(s)
- Mahmut Mijit
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kamakshi Sishtla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gabriella D. Hartman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Timothy W. Corson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Melvin and Bren 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
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark R. Kelley
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
19
|
Sarwar A, Zhu M, Su Q, Zhu Z, Yang T, Chen Y, Peng X, Zhang Y. Targeting mitochondrial dysfunctions in pancreatic cancer evokes new therapeutic opportunities. Crit Rev Oncol Hematol 2022; 180:103858. [DOI: 10.1016/j.critrevonc.2022.103858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/07/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
|
20
|
Wang C, Zhou W, Liu Y, Xu Y, Zhang X, Jiang C, Jiang M, Cao X. Nuclear translocation of RIG-I promotes cellular apoptosis. J Autoimmun 2022; 130:102840. [DOI: 10.1016/j.jaut.2022.102840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022]
|
21
|
Guo N, Chen Y, Zhang Y, Deng Y, Zeng F, Li X. Potential Role of APEX1 During Ferroptosis. Front Oncol 2022; 12:798304. [PMID: 35311089 PMCID: PMC8927806 DOI: 10.3389/fonc.2022.798304] [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: 10/20/2021] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
Ferroptosis is a recently discovered category of programmed cell death. It is much different from other types of cell death such as apoptosis, necrosis and autophagy. The main pathological feature of ferroptosis is the accumulation of iron-dependent lipid peroxidation. The typical changes in the morphological features of ferroptosis include cell volume shrinkage and increased mitochondrial membrane area. The mechanisms of ferroptosis may be mainly related to lipid peroxidation accumulation, imbalance in amino acid antioxidant system, and disturbance of iron metabolism. Besides, hypoxia-inducible factor (HIF), nuclear factor-E2-related factor 2 (Nrf2), and p53 pathway have been demonstrated to be involved in ferroptosis. At present, the molecular mechanisms of ferroptosis pathway are still unmapped. In this review, an outlook has been put forward about the crucial role of apurinic/apyrimidinic endodeoxyribonuclease 1 (APEX1) in the regulation of ferroptosis. APEX1 plays an important role in the regulation of intracellular redox balance and can be used as a potential inhibitor of ferroptotic cell death. Bioinformatics analysis indicated that the mRNA level of APEX1 is decreased in cases of ferroptosis triggered by erastin. Besides, it was found that there was a significant correlation between APEX1 and genes in the ferroptosis pathway. We have discussed the possibility to employ APEX1 inducers or inhibitors in the regulation of ferroptosis as a new strategy for the treatment of various human diseases.
Collapse
Affiliation(s)
- Nan Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Yan Chen
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhong Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Yonghao Deng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Kalinin S, Malkova A, Sharonova T, Sharoyko V, Bunev A, Supuran CT, Krasavin M. Carbonic Anhydrase IX Inhibitors as Candidates for Combination Therapy of Solid Tumors. Int J Mol Sci 2021; 22:13405. [PMID: 34948200 PMCID: PMC8705727 DOI: 10.3390/ijms222413405] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Combination therapy is becoming imperative for the treatment of many cancers, as it provides a higher chance of avoiding drug resistance and tumor recurrence. Among the resistance-conferring factors, the tumor microenvironment plays a major role, and therefore, represents a viable target for adjuvant therapeutic agents. Thus, hypoxia and extracellular acidosis are known to select for the most aggressive and resilient phenotypes and build poorly responsive regions of the tumor mass. Carbonic anhydrase (CA, EC 4.2.1.1) IX isoform is a surficial zinc metalloenzyme that is proven to play a central role in regulating intra and extracellular pH, as well as modulating invasion and metastasis processes. With its strong association and distribution in various tumor tissues and well-known druggability, this protein holds great promise as a target to pharmacologically interfere with the tumor microenvironment by using drug combination regimens. In the present review, we summarized recent publications revealing the potential of CA IX inhibitors to intensify cancer chemotherapy and overcome drug resistance in preclinical settings.
Collapse
Affiliation(s)
- Stanislav Kalinin
- Institute of Chemistry, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.M.); (T.S.); (V.S.); (M.K.)
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
- Medicinal Chemistry Center, Togliatti State University, 445020 Togliatti, Russia;
| | - Anna Malkova
- Institute of Chemistry, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.M.); (T.S.); (V.S.); (M.K.)
| | - Tatiana Sharonova
- Institute of Chemistry, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.M.); (T.S.); (V.S.); (M.K.)
| | - Vladimir Sharoyko
- Institute of Chemistry, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.M.); (T.S.); (V.S.); (M.K.)
- Medicinal Chemistry Center, Togliatti State University, 445020 Togliatti, Russia;
| | - Alexander Bunev
- Medicinal Chemistry Center, Togliatti State University, 445020 Togliatti, Russia;
| | - Claudiu T. Supuran
- Neurofarba Department, Universita degli Studi di Firenze, 50019 Florence, Italy;
| | - Mikhail Krasavin
- Institute of Chemistry, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.M.); (T.S.); (V.S.); (M.K.)
| |
Collapse
|
23
|
CpG Island Methylator Phenotype Modulates the Immune Response of the Tumor Microenvironment and Influences the Prognosis of Pancreatic Cancer Patients. JOURNAL OF ONCOLOGY 2021; 2021:2715694. [PMID: 34876903 PMCID: PMC8645373 DOI: 10.1155/2021/2715694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/22/2021] [Indexed: 12/25/2022]
Abstract
Background CpG island methylator phenotype (CIMP), featured with concurrent and widespread hypermethylation of a cluster of CpGs, has been reported to play an important role in carcinogenesis. Limited studies have investigated the role of CIMP in pancreatic cancer (PC). The aim of this study was to explore the CIMP in PC patients and its impact on the immune response of the tumor microenvironment and prognosis. Methods DNA methylation, somatic mutation, mRNA, and corresponding clinical data of PC patients were downloaded from TCGA (184 patients) and the ICGC (264 patients). Univariate and multivariate regression analyses were used to identify prognosis-related CpGs. Consensus clustering analysis was used for identification of the CIMP in PC patients. ESTIMATE and CIBORORT were used for estimation of the tumor microenvironment (TME) in PC patients. Results In the TCGA PC cohort, 22,450 differential CpGs, including 12,937 hypermethylated CpGs and 9,513 hypomethylated CpGs, were identified between 184 PC patients and 10 normal controls. Univariate and multivariate Cox analysis further screened out 72 OS-related CpGs, and three distinct CIMP groups with distinctly different prognosis and molecular features, including the CIMP-L subgroup, CIMP-M subgroup, and CIMP-H subgroup, were identified based on unsupervised consensus clustering analysis of these CpGs. Patients of the CIMP-H subgroup had poorer OS and RFS, while patients of the CIMP-L subgroup had better OS and RFS. The CIMP status was also an independent prognostic factor for OS and PFS. In molecular features, significantly higher somatic mutation burden and tumor mutational burden were found in patients of the CIMP-H subgroup compared to those of the CIMP-L subgroup. Besides, lower stromal score, immune score, and higher cancer stemness indices and tumor purity were also found in patients of the CIMP-H subgroup compared to those of the CIMP-L subgroup. Correspondingly, significant total T cells, total B cells, CD8 T cells, memory CD4 T cells, and higher regulatory T cells were found in patients of the CIMP-H subgroup. Moreover, significantly lower expression of immune checkpoint genes, such as PD-1, CTLA4, CD86, VTCN1, and LAG-3, was also found in patients of the CIMP-H subgroup compared to those of the CIMP-L subgroup. In the end, we validated the CIMP status in PC patients of the ICGC dataset. Conclusion The CIMP may modulate the immune response of the tumor microenvironment and influence the prognosis of pancreatic cancer patients, which may help to make an assertion to provide specific and efficient treatment options for patients of different subtypes.
Collapse
|
24
|
Campillo-Gimenez L, Rios-Covian D, Rivera-Nieves J, Kiyono H, Chu H, Ernst PB. Microbial-Driven Immunological Memory and Its Potential Role in Microbiome Editing for the Prevention of Colorectal Cancer. Front Cell Infect Microbiol 2021; 11:752304. [PMID: 34869061 PMCID: PMC8633303 DOI: 10.3389/fcimb.2021.752304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Over the last several years, many advances have been made in understanding the role of bacteria in the pathogenesis of gastrointestinal cancers. Beginning with Helicobacter pylori being recognized as the first bacterial carcinogen and the causative agent of most gastric cancers, more recent studies have examined the role of enteric microbes in colorectal cancer. In the digestive tract, these communities are numerous and have a complex interrelationship with local immune/inflammatory responses that impact the health of the host. As modifying the microbiome in the stomach has decreased the risk of gastric cancer, modifying the distal microbiome may decrease the risk of colorectal cancers. To date, very few studies have considered the notion that mucosal lymphocyte-dependent immune memory may confound attempts to change the microbial components in these communities. The goal of this review is to consider some of the factors impacting host-microbial interactions that affect colorectal cancer and raise questions about how immune memory responses to the local microbial consortium affect any attempt to modify the composition of the intestinal microbiome.
Collapse
Affiliation(s)
- Laure Campillo-Gimenez
- Department of Pathology, University of California San Diego, San Diego, CA, United States
| | - David Rios-Covian
- Department of Pathology, University of California San Diego, San Diego, CA, United States
| | - Jesus Rivera-Nieves
- Department of Medicine, Division of Gastroenterology, University of California San Diego, San Diego, CA, United States
- San Diego Veterans Affairs (VA) Medical Center, San Diego, CA, United States
| | - Hiroshi Kiyono
- Department of Medicine, Division of Gastroenterology, University of California San Diego, San Diego, CA, United States
- CU-UCSD, Center for Mucosal Immunology, Allergy and Vaccine Development, University of California San Diego, San Diego, CA, United States
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan
| | - Hiutung Chu
- Department of Pathology, University of California San Diego, San Diego, CA, United States
- CU-UCSD, Center for Mucosal Immunology, Allergy and Vaccine Development, University of California San Diego, San Diego, CA, United States
| | - Peter B. Ernst
- Department of Pathology, University of California San Diego, San Diego, CA, United States
- San Diego Veterans Affairs (VA) Medical Center, San Diego, CA, United States
- CU-UCSD, Center for Mucosal Immunology, Allergy and Vaccine Development, University of California San Diego, San Diego, CA, United States
- Division of Comparative Pathology and Medicine, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
25
|
Elbanna M, Chowdhury NN, Rhome R, Fishel ML. Clinical and Preclinical Outcomes of Combining Targeted Therapy With Radiotherapy. Front Oncol 2021; 11:749496. [PMID: 34733787 PMCID: PMC8558533 DOI: 10.3389/fonc.2021.749496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
In the era of precision medicine, radiation medicine is currently focused on the precise delivery of highly conformal radiation treatments. However, the tremendous developments in targeted therapy are yet to fulfill their full promise and arguably have the potential to dramatically enhance the radiation therapeutic ratio. The increased ability to molecularly profile tumors both at diagnosis and at relapse and the co-incident progress in the field of radiogenomics could potentially pave the way for a more personalized approach to radiation treatment in contrast to the current ‘‘one size fits all’’ paradigm. Few clinical trials to date have shown an improved clinical outcome when combining targeted agents with radiation therapy, however, most have failed to show benefit, which is arguably due to limited preclinical data. Several key molecular pathways could theoretically enhance therapeutic effect of radiation when rationally targeted either by directly enhancing tumor cell kill or indirectly through the abscopal effect of radiation when combined with novel immunotherapies. The timing of combining molecular targeted therapy with radiation is also important to determine and could greatly affect the outcome depending on which pathway is being inhibited.
Collapse
Affiliation(s)
- May Elbanna
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nayela N Chowdhury
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ryan Rhome
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Melissa L Fishel
- Indiana University Simon Comprehensive Cancer Center, 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 Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
26
|
Oliveira TT, Fontes-Dantas FL, de Medeiros Oliveira RK, Pinheiro DML, Coutinho LG, da Silva VL, de Souza SJ, Agnez-Lima LF. Chemical Inhibition of Apurinic-Apyrimidinic Endonuclease 1 Redox and DNA Repair Functions Affects the Inflammatory Response via Different but Overlapping Mechanisms. Front Cell Dev Biol 2021; 9:731588. [PMID: 34616737 PMCID: PMC8488223 DOI: 10.3389/fcell.2021.731588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/27/2021] [Indexed: 01/21/2023] Open
Abstract
The presence of oxidized DNA lesions, such as 7,8-dihydro-8-oxoguanine (8-oxoG) and apurinic/apyrimidinic sites (AP sites), has been described as epigenetic signals that are involved in gene expression control. In mammals, Apurinic-apyrimidinic endonuclease 1/Redox factor-1 (APE1/Ref-1) is the main AP endonuclease of the base excision repair (BER) pathway and is involved in active demethylation processes. In addition, APE1/Ref-1, through its redox function, regulates several transcriptional factors. However, the transcriptional control targets of each APE1 function are not completely known. In this study, a transcriptomic approach was used to investigate the effects of chemical inhibition of APE1/Ref-1 redox or DNA repair functions by E3330 or methoxyamine (MX) in an inflammatory cellular model. Under lipopolysaccharide (LPS) stimulation, both E3330 and MX reduced the expression of some cytokines and chemokines. Interestingly, E3330 treatment reduced cell viability after 48 h of the treatment. Genes related to inflammatory response and mitochondrial processes were downregulated in both treatments. In the E3330 treatment, RNA processing and ribosome biogenesis genes were downregulated, while they were upregulated in the MX treatment. Furthermore, in the E3330 treatment, the cellular stress response was the main upregulated process, while the cellular macromolecule metabolic process was observed in MX-upregulated genes. Nuclear respiratory factor 1 (NRF1) was predicted to be a master regulator of the downregulated genes in both treatments, while the ETS transcription factor ELK1 (ELK1) was predicted to be a master regulator only for E3330 treatment. Decreased expression of ELK1 and its target genes and a reduced 28S/18S ratio were observed, suggesting impaired rRNA processing. In addition, both redox and repair functions can affect the expression of NRF1 and GABPA target genes. The master regulators predicted for upregulated genes were YY1 and FLI1 for the E3330 and MX treatments, respectively. In summary, the chemical inhibition of APE1/Ref-1 affects gene expression regulated mainly by transcriptional factors of the ETS family, showing partial overlap of APE1 redox and DNA repair functions, suggesting that these activities are not entirely independent. This work provides a new perspective on the interaction between APE1 redox and DNA repair activity in inflammatory response modulation and transcription.
Collapse
Affiliation(s)
- Thais Teixeira Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil
| | - Fabrícia Lima Fontes-Dantas
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil
| | | | | | - Leonam Gomes Coutinho
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil.,Instituto Federal de Educação Tecnológica do Rio Grande do Norte, IFRN, São Paulo do Potengi, Brazil
| | - Vandeclecio Lira da Silva
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal, Brazil.,Bioinformatics Multidisciplinary Environment (BioME), IMD, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Sandro José de Souza
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal, Brazil.,Bioinformatics Multidisciplinary Environment (BioME), IMD, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | | |
Collapse
|
27
|
Beyond the Double-Strand Breaks: The Role of DNA Repair Proteins in Cancer Stem-Cell Regulation. Cancers (Basel) 2021; 13:cancers13194818. [PMID: 34638302 PMCID: PMC8508278 DOI: 10.3390/cancers13194818] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer stem cells (CSCs) are a tumor cell population maintaining tumor growth and promoting tumor relapse if not wholly eradicated during treatment. CSCs are often equipped with molecular mechanisms making them resistant to conventional anti-cancer therapies whose curative potential depends on DNA damage-induced cell death. An elevated expression of some key DNA repair proteins is one of such defense mechanisms. However, new research reveals that the role of critical DNA repair proteins is extending far beyond the DNA repair mechanisms. This review discusses the diverse biological functions of DNA repair proteins in CSC maintenance and the adaptation to replication and oxidative stress, anti-cancer immune response, epigenetic reprogramming, and intracellular signaling mechanisms. It also provides an overview of their potential therapeutic targeting. Abstract Cancer stem cells (CSCs) are pluripotent and highly tumorigenic cells that can re-populate a tumor and cause relapses even after initially successful therapy. As with tissue stem cells, CSCs possess enhanced DNA repair mechanisms. An active DNA damage response alleviates the increased oxidative and replicative stress and leads to therapy resistance. On the other hand, mutations in DNA repair genes cause genomic instability, therefore driving tumor evolution and developing highly aggressive CSC phenotypes. However, the role of DNA repair proteins in CSCs extends beyond the level of DNA damage. In recent years, more and more studies have reported the unexpected role of DNA repair proteins in the regulation of transcription, CSC signaling pathways, intracellular levels of reactive oxygen species (ROS), and epithelial–mesenchymal transition (EMT). Moreover, DNA damage signaling plays an essential role in the immune response towards tumor cells. Due to its high importance for the CSC phenotype and treatment resistance, the DNA damage response is a promising target for individualized therapies. Furthermore, understanding the dependence of CSC on DNA repair pathways can be therapeutically exploited to induce synthetic lethality and sensitize CSCs to anti-cancer therapies. This review discusses the different roles of DNA repair proteins in CSC maintenance and their potential as therapeutic targets.
Collapse
|
28
|
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: 3.3] [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
|
29
|
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: 5.8] [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
|
30
|
Estaras M, Gonzalez A. Modulation of cell physiology under hypoxia in pancreatic cancer. World J Gastroenterol 2021; 27:4582-4602. [PMID: 34366624 PMCID: PMC8326256 DOI: 10.3748/wjg.v27.i28.4582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/28/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
In solid tumors, the development of vasculature is, to some extent, slower than the proliferation of the different types of cells that form the tissue, both cancer and stroma cells. As a consequence, the oxygen availability is compromised and the tissue evolves toward a condition of hypoxia. The presence of hypoxia is variable depending on where the cells are localized, being less extreme at the periphery of the tumor and more severe in areas located deep within the tumor mass. Surprisingly, the cells do not die. Intracellular pathways that are critical for cell fate such as endoplasmic reticulum stress, apoptosis, autophagy, and others are all involved in cellular responses to the low oxygen availability and are orchestrated by hypoxia-inducible factor. Oxidative stress and inflammation are critical conditions that develop under hypoxia. Together with changes in cellular bioenergetics, all contribute to cell survival. Moreover, cell-to-cell interaction is established within the tumor such that cancer cells and the microenvironment maintain a bidirectional communication. Additionally, the release of extracellular vesicles, or exosomes, represents short and long loops that can convey important information regarding invasion and metastasis. As a result, the tumor grows and its malignancy increases. Currently, one of the most lethal tumors is pancreatic cancer. This paper reviews the most recent advances in the knowledge of how cells grow in a pancreatic tumor by adapting to hypoxia. Unmasking the physiological processes that help the tumor increase its size and their regulation will be of major relevance for the treatment of this deadly tumor.
Collapse
Affiliation(s)
- Matias Estaras
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres 10003, Spain
| | - Antonio Gonzalez
- Department of Physiology, Cell Biology and Communication Research Group, University of Extremadura, Caceres 10003, Spain
| |
Collapse
|
31
|
Fu X, Feng Y, Shao B, Zhang Y. Taxifolin Protects Dental Pulp Stem Cells under Hypoxia and Inflammation Conditions. Cell Transplant 2021; 30:9636897211034452. [PMID: 34292054 PMCID: PMC8312191 DOI: 10.1177/09636897211034452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background: Dental pulp stem cells (DPSCs) are a unique source for future clinical application in dentistry such as periodontology or endodontics. However, DPSCs are prone to apoptosis under abnormal conditions. Taxifolin is a natural flavonoid and possesses many pharmacological activities including anti-hypoxic and anti-inflammatory. We aimed to elucidate the mechanisms of taxifolin protects DPSC under hypoxia and inflammatory conditions. Methods: DPSCs from human dental pulp tissue was purchased from Lonza (cat. no. PT-5025. Basel, Switzerland)) and identified by DPSC’s biomarkers. DPSC differentiation in vitro following the manufacturers’ instructions. ARS staining and Oil red staining verify the efficiency of differentiation in vitro after 2 weeks. The changes of various genes and proteins were identified by Q-PCR and western-blot, respectively. Cell viability was determined by the CCK-8 method, while apoptosis was determined by Annexin V/PI staining. Results: DPSC differentiation in vitro shows that hypoxia and TNF-α synergistically inhibit the survival and osteogenesis of DPSCs. A final concentration of 10 μM Taxifolin can significantly reduce the apoptosis of DPSCs under inflammation and hypoxia conditions. Taxifolin substantially increases carbonic anhydrase IX (CA9) expression but not HIF1a, and inhibitions of CA9 expression nullify the protective role of taxifolin under hypoxia and inflammatory condition. Conclusion: Taxifolin significantly increased the expression of CA9 when it inhibits DPSC apoptosis and taxifolin synergistically to protect DPSCs against apoptosis with CA9 under hypoxia and inflammatory conditions. Taxifolin can be used as a potential drug for clinical treatment of DPSC-related diseases.
Collapse
Affiliation(s)
- Xiaohui Fu
- Department of General Dentistry, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, P.R. China
| | - Yimiao Feng
- Department of Orthodontics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Bingyi Shao
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Yubei District, Chongqing, P. R. China
| | - Yanzhen Zhang
- Department of General Dentistry, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, P.R. China
| |
Collapse
|
32
|
Shahmoradi Ghahe S, Kosicki K, Wojewódzka M, Majchrzak BA, Fogtman A, Iwanicka-Nowicka R, Ciuba A, Koblowska M, Kruszewski M, Tudek B, Speina E. Increased DNA repair capacity augments resistance of glioblastoma cells to photodynamic therapy. DNA Repair (Amst) 2021; 104:103136. [PMID: 34044336 DOI: 10.1016/j.dnarep.2021.103136] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/15/2021] [Indexed: 12/21/2022]
Abstract
Photodynamic therapy (PDT) is a clinically approved cancer therapy of low invasiveness. The therapeutic procedure involves administering a photosensitizing drug (PS), which is then activated with monochromatic light of a specific wavelength. The photochemical reaction produces highly toxic oxygen species. The development of resistance to PDT in some cancer cells is its main limitation. Several mechanisms are known to be involved in the development of cellular defense against cytotoxic effects of PDT, including activation of antioxidant enzymes, drug efflux pumps, degradation of PS, and overexpression of protein chaperons. Another putative factor that plays an important role in the development of resistance of cancer cells to PDT seems to be DNA repair; however, it has not been well studied so far. To explore the role of DNA repair and other potential novel mechanisms associated with the resistance to PDT in the glioblastoma cells, cells stably resistant to PDT were isolated from PDT sensitive cells following repetitive PDT cycles. Duly characterization of isolated PDT-resistant glioblastoma revealed that the resistance to PDT might be a consequence of several mechanisms, including higher repair efficiency of oxidative DNA damage and repair of DNA breaks. Higher activity of APE1 endonuclease and increased expression and activation of DNA damage kinase ATM was demonstrated in the U-87 MGR cell line, suggesting and proving that they are good targets for sensitization of resistant cells to PDT.
Collapse
Affiliation(s)
- Somayeh Shahmoradi Ghahe
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland; Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Konrad Kosicki
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Maria Wojewódzka
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195, Warsaw, Poland
| | - Bartosz A Majchrzak
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Anna Fogtman
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland; Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland; Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Agata Ciuba
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Marta Koblowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland; Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195, Warsaw, Poland; Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090, Lublin, Poland
| | - Barbara Tudek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland; Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Elżbieta Speina
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland.
| |
Collapse
|
33
|
Rahnamay Farnood P, Danesh Pazhooh R, Asemi Z, Yousefi B. DNA damage response and repair in pancreatic cancer development and therapy. DNA Repair (Amst) 2021; 103:103116. [PMID: 33882393 DOI: 10.1016/j.dnarep.2021.103116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer (PC) is among fatal malignancies, with a dismal prognosis and a low survival rate of 5-10%. In both sporadic and inherited PC, gene alterations, such as BRCA1/2, PALB2, and ATM, can occur frequently. Currently, surgery, chemo- and radio-therapy are the most common therapeutic strategies for treating this cancer. DNA damage response (DDR) establishes multiple pathways that eliminate DNA damage sites to maintain genomic integrity. Various types of cancers and age-related diseases are associated with DDR machinery defects. According to the severity of the damage, DDR pathways respond appropriately to lesions through repairing damage, arresting the cell cycle, or apoptosis. Recently, novel agents, particularly those targeting DDR pathways, are being utilized to improve the response of many cancers to chemotherapy and radiotherapy. In this paper, we briefly reviewed DDR processes and their components, including DDR sensors, DDR mediators, and DDR transducers in the progression, prognosis, and treatment of PC.
Collapse
Affiliation(s)
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
34
|
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: 68] [Impact Index Per Article: 17.0] [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
|
35
|
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: 1.3] [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
|
36
|
Hu J, Wang Y, Yuan Y. Inhibitors of APE1 redox function effectively inhibit γ-herpesvirus replication in vitro and in vivo. Antiviral Res 2020; 185:104985. [PMID: 33271272 DOI: 10.1016/j.antiviral.2020.104985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
APE1 is a multi-functional protein with a redox function in its N-terminal domain and an apurinic/apyrimidinic endonuclease activity in the C-terminal domain. APE1 redox function plays an important role in regulating cell proliferation and survival through activating specific transcriptional activators. APE1 redox function is also found to be associated with some cancer occurrence. In this study, we demonstrated that APE1 redox function is essential for Epstein-Barr virus (EBV) lytic replication as the silencing of APE1 expression or treatment with APE1 redox inhibitors C10 and E3330 can inhibit EBV lytic replication and virion production. Furthermore, C10 and E3330 also inhibit MHV-68 replication in vitro and in vivo. C10 and E3330 were able to significantly reduce the loss of pulmonary alveoli and thickening of alveolar septa in mice caused by MHV-68 infection. Altogether, (i) APE1 redox function is validated as a new antiviral target; (ii) APE1 redox inhibitors, especially C10, have potentials to be used for the treatment of γ-herpesvirus infection and associated diseases; (iii) MHV-68 is validated to be a surrogate for the study of the pathogenesis and therapy of EBV and KSHV infection in vivo.
Collapse
Affiliation(s)
- Jiayuan Hu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Wang
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yan Yuan
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
The redox function of apurinic/apyrimidinic endonuclease 1 as key modulator in photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 211:111992. [DOI: 10.1016/j.jphotobiol.2020.111992] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 01/04/2023]
|
38
|
Deng X, Zhen P, Niu X, Dai Y, Wang Y, Zhou M. APE1 promotes proliferation and migration of cutaneous squamous cell carcinoma. J Dermatol Sci 2020; 100:67-74. [PMID: 32951990 DOI: 10.1016/j.jdermsci.2020.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/09/2020] [Accepted: 08/23/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Human Apurinic/Apyrimidinic Endonuclease 1 (APE1/REF-1/HAP1) is a multifunction protein involved in the progression of cancer. But the role of APE1 in cutaneous squamous cell carcinoma (cSCC) is unclear. OBJECTIVE This study is aimed to investigate the basic modulatory mechanism of APE1 in cSCC development and offer a novel potential target for clinical treatment. METHODS The expression of APE1 in cSCC tissues was detected by western blot and immunohistochemistry (IHC) staining. The function of APE1 and miR-27a in cSCC cells was investigated by cell counting kit-8 (CCK-8) assays, colony formation assays and transwell migration assays. Western blot was used to determine the expression of APE1 in cSCC and epithelial-mesenchymal transition (EMT) markers in HSC-1 and HSC-5 cells with APE1 knockdown or overexpression. Double luciferase reporter assays were performed to confirm the interaction of miR-27a and APE1. RESULTS We identified that APE1 was significantly upregulated in human cSCC tissues and cSCC cells and its overexpression promoted cell proliferation, migration and the expression of EMT markers in cSCC cells. Mechanistically, miR-27a was predicted and confirmed as the upstream of APE1. Its downregulation also enhanced the proliferation and migration of cSCC cells. Rescue experiments demonstrated that restoration of APE1 expression significantly abolished the inhibition of cell proliferation and migration mediated by miR-27a. CONCLUSION As a direct gene of miR-27a, APE1 improved cell proliferation and migration to promote the progression of cSCC, which could be considered as a potential therapeutic target for cSCC treatment.
Collapse
Affiliation(s)
- Xuyi Deng
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangdong, China
| | - Peilin Zhen
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Guangdong, China
| | - Xinli Niu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangdong, China
| | - Yu Dai
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - Yinghui Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangdong, China; Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Guangdong, China.
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangdong, China.
| |
Collapse
|
39
|
Strapcova S, Takacova M, Csaderova L, Martinelli P, Lukacikova L, Gal V, Kopacek J, Svastova E. Clinical and Pre-Clinical Evidence of Carbonic Anhydrase IX in Pancreatic Cancer and Its High Expression in Pre-Cancerous Lesions. Cancers (Basel) 2020; 12:E2005. [PMID: 32707920 PMCID: PMC7464147 DOI: 10.3390/cancers12082005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Hypoxia is a common phenomenon that occurs in most solid tumors. Regardless of tumor origin, the evolution of a hypoxia-adapted phenotype is critical for invasive cancer development. Pancreatic ductal adenocarcinoma is also characterized by hypoxia, desmoplasia, and the presence of necrosis, predicting poor outcome. Carbonic anhydrase IX (CAIX) is one of the most strict hypoxia regulated genes which plays a key role in the adaptation of cancer cells to hypoxia and acidosis. Here, we summarize clinical data showing that CAIX expression is associated with tumor necrosis, vascularization, expression of Frizzled-1, mucins, or proteins involved in glycolysis, and inevitably, poor prognosis of pancreatic cancer patients. We also describe the transcriptional regulation of CAIX in relation to signaling pathways activated in pancreatic cancers. A large part deals with the preclinical evidence supporting the relevance of CAIX in processes leading to the aggressive behavior of pancreatic tumors. Furthermore, we focus on CAIX occurrence in pre-cancerous lesions, and for the first time, we describe CAIX expression within intraductal papillary mucinous neoplasia. Our review concludes with a detailed account of clinical trials implicating that treatment consisting of conventionally used therapies combined with CAIX targeting could result in an improved anti-cancer response in pancreatic cancer patients.
Collapse
Affiliation(s)
- Sabina Strapcova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Martina Takacova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Lucia Csaderova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Paola Martinelli
- Institute of Cancer Research, Clinic of Internal Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
- Cancer Cell Signaling, Boehringer-Ingelheim RCV Vienna, A-1121 Vienna, Austria
| | - Lubomira Lukacikova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Viliam Gal
- Alpha Medical Pathology, Ruzinovska 6, 82606 Bratislava, Slovakia;
| | - Juraj Kopacek
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Eliska Svastova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| |
Collapse
|
40
|
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: 24] [Impact Index Per Article: 4.8] [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
|
41
|
Dietary walnut as food factor to rescue from NSAID-induced gastrointestinal mucosal damages. Arch Biochem Biophys 2020; 689:108466. [PMID: 32590067 DOI: 10.1016/j.abb.2020.108466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/08/2020] [Accepted: 06/07/2020] [Indexed: 01/22/2023]
Abstract
Nuclear factor erythroid-derived 2-like 2 (Nrf-2) is transcription factor implicated in the antioxidant response element-mediated induction of endogenous antioxidant enzyme such as heme oxygenase-1 (HO-1), glutamate-cysteine ligase, and NAD(P)H quinone dehydrogenase 1, among which HO-1 is an enzyme catalyzing the degradation of heme.producing biliverdin, ferrous iron, and carbon monoxide. In the stomach, as much as regulating gastric acid secretions, well-coordinated establishment of defense system stands for maintaining gastric integrity. In previous study, author et al. for the first time discovered HO-1 induction was critical in affording faithful gastric defense against various irritants including Helicobacter pylori infection, stress, alcohol, non-steroidal anti-inflammatory drugs (NSAIDs), aspirin, and toxic bile acids. In this review article, we can add the novel evidence that dietary walnut intake can be reliable way to rescue from NSAIDs-induced gastrointestinal damages via the induction of HO-1 transcribed with Nrf-2 through specific inactivation of Keap-1. From molecular exploration to translational animal model of indomethacin-induced gastrointestinal damages, significant induction of HO-1 contributed to rescuing from damages. In addition to HO-1 induction action relevant to walnut, we added the description the general actions of walnut extracts or dietary intake of walnut regarding cytoprotection and why we have focused on to NSAID damages.
Collapse
|
42
|
Choi YD, Jung JY, Baek M, Khan S, Song PI, Ryu S, Koo JY, Chauhan SC, Tsin A, Choi C, Kim WJ, Kim M. APE1 Promotes Pancreatic Cancer Proliferation through GFRα1/Src/ERK Axis-Cascade Signaling in Response to GDNF. Int J Mol Sci 2020; 21:E3586. [PMID: 32438692 PMCID: PMC7279477 DOI: 10.3390/ijms21103586] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer is the worst exocrine gastrointestinal cancer leading to the highest mortality. Recent studies reported that aberrant expression of apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) is involved in uncontrolled cell growth. However, the molecular mechanism of APE1 biological role remains unrevealed in pancreatic cancer progression. Here, we demonstrate that APE1 accelerates pancreatic cancer cell proliferation through glial cell line-derived neurotrophic factor (GDNF)/glial factor receptor α1 (GFRα1)/Src/ERK axis-cascade signaling. The proliferation of endogenous APE1 expressed-MIA PaCa-2, a human pancreatic carcinoma cell line, was increased by treatment with GDNF, a ligand of GFRα1. Either of downregulated APE1 or GFRα1 expression using small interference RNA (siRNA) inhibited GDNF-induced cancer cell proliferation. The MEK-1 inhibitor PD98059 decreased GDNF-induced MIA PaCa-2 cell proliferation. Src inactivation by either its siRNA or Src inhibitor decreased ERK-phosphorylation in response to GDNF in MIA PaCa-2 cells. Overexpression of GFRα1 in APE1-deficient MIA PaCa-2 cells activated the phosphorylation of Src and ERK. The expression of both APE1 and GFRα1 was gradually increased as progressing pancreatic cancer grades. Our results highlight a critical role for APE1 in GDNF-induced pancreatic cancer cell proliferation through APE1/GFRα1/Src/ERK axis-cascade signaling and provide evidence for future potential therapeutic drug targets for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yoo-Duk Choi
- Department of Pathology, Chonnam National University Medical School, Gwangju 61186, Korea; (Y.-D.C.); (J.-Y.K.)
| | - Ji-Yeon Jung
- Dental Science Research Institute, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Minwoo Baek
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN 55812, USA;
| | - Sheema Khan
- Department of Immunology & Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.K.); (S.C.C.)
| | - Peter I. Song
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (P.I.S.); (A.T.)
| | - Sunhyo Ryu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA;
| | - Joo-Yeon Koo
- Department of Pathology, Chonnam National University Medical School, Gwangju 61186, Korea; (Y.-D.C.); (J.-Y.K.)
| | - Subhash C. Chauhan
- Department of Immunology & Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.K.); (S.C.C.)
| | - Andrew Tsin
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (P.I.S.); (A.T.)
| | - Chan Choi
- Department of Pathology, Chonnam National University Hwasun Hospital, Hwasun 58128, Korea;
| | - Won Jae Kim
- Dental Science Research Institute, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Mihwa Kim
- Department of Pathology, Chonnam National University Medical School, Gwangju 61186, Korea; (Y.-D.C.); (J.-Y.K.)
- Dental Science Research Institute, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (P.I.S.); (A.T.)
| |
Collapse
|
43
|
Santos HBDP, Morais EFD, Cavalcante RB, Nogueira RLM, Nonaka CFW, Souza LBD, Freitas RDA. Immunoexpression of DNA base excision repair and nucleotide excision repair proteins in ameloblastomas, syndromic and non-syndromic odontogenic keratocysts and dentigerous cysts. Arch Oral Biol 2019; 110:104627. [PMID: 31862643 DOI: 10.1016/j.archoralbio.2019.104627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To evaluate the immunoexpression of DNA base excision repair (BER) [apurinic/apyrimidinic endonuclease 1 (APE-1), X-ray repair cross complementing 1 (XRCC-1)] and nucleotide excision repair (NER) [xeroderma pigmentosum complementation group (XPF)] proteins in benign epithelial odontogenic lesions with different biological behaviors. DESIGN Thirty solid ameloblastomas, 30 non-syndromic odontogenic keratocysts (NSOKCs), 29 syndromic odontogenic keratocysts (SKOCs), 30 dentigerous cysts (DCs) and 20 dental follicles (DFs) were evaluated quantitatively for APE-1, XRCC-1 and XPF through immunohistochemistry. RESULTS Nuclear expression of APE-1 was significantly higher in NSOKCs, SOKCs, and ameloblastomas in comparison to DCs (p < 0.001). Nuclear expression of XRCC-1 was higher in NSOKCs and SOKCs than in DCs (p < 0.05). At the nuclear level, XPF expression was higher in NSOKCs and SOKCs than in DCs and ameloblastomas (p < 0.05). A statistically significant higher expression of APE-1 (nuclear), XRCC-1 (nuclear), and XPF (nuclear and cytoplasmic) was found in all odontogenic lesion samples as compared to DFs (p < 0.05). For all lesions, there was a positive correlation between nuclear expression of APE-1 and XRCC-1 or XPF (p < 0.05). CONCLUSIONS Our results suggest a potential involvement of APE-1, XRCC-1 and XPF proteins in the pathogenesis of benign epithelial odontogenic lesions, especially in those with more aggressive biological behavior, such as ameloblastomas, NSOKCs, and SOKCs. We also showed that the expression of APE-1 was positively correlated with the nuclear expression of XRCC-1 and XPF, which may suggest an interaction between the BER and NER pathways in all odontogenic lesions studied herein.
Collapse
|
44
|
Sun HR, Wang S, Yan SC, Zhang Y, Nelson PJ, Jia HL, Qin LX, Dong QZ. Therapeutic Strategies Targeting Cancer Stem Cells and Their Microenvironment. Front Oncol 2019; 9:1104. [PMID: 31709180 PMCID: PMC6821685 DOI: 10.3389/fonc.2019.01104] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) have been demonstrated in a variety of tumors and are thought to act as a clonogenic core for the genesis of new tumor growth. This small subpopulation of cancer cells has been proposed to help drive tumorigenesis, metastasis, recurrence and conventional therapy resistance. CSCs show self-renewal and flexible clonogenic properties and help define specific tumor microenvironments (TME). The interaction between CSCs and TME is thought to function as a dynamic support system that fosters the generation and maintenance of CSCs. Investigation of the interaction between CSCs and the TME is shedding light on the biologic mechanisms underlying the process of tumor malignancy, metastasis, and therapy resistance. We summarize recent advances in CSC biology and their environment, and discuss the challenges and future strategies for targeting this biology as a new therapeutic approach.
Collapse
Affiliation(s)
- Hao-Ran Sun
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shun Wang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Can Yan
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter J. Nelson
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Hu-Liang Jia
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong-Zhu Dong
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
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: 4.3] [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
|
46
|
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: 4.3] [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
|
47
|
Li H, Zhong C, Wang Q, Chen W, Yuan Y. Curcumin is an APE1 redox inhibitor and exhibits an antiviral activity against KSHV replication and pathogenesis. Antiviral Res 2019; 167:98-103. [PMID: 31034848 DOI: 10.1016/j.antiviral.2019.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 01/06/2023]
Abstract
Curcumin, a polyphenol, is the main bioactive compound in dietary spice turmeric curcuma longa. It possesses anti-inflammatory, anti-oxidant and anti-neoplastic properties and shows potentials in treating or preventing particular diseases such as oxidative and inflammatory conditions, metabolic syndrome, arthritis, anxiety, hyperlipidemia and cancers. The diverse range and potential health beneficial effects has generated enthusiasm leading to intensive investigation into the phytochemical. However, a concern has been also raised if curcumin has a promiscuous bioassay profile and is a Pan-Assay INterference compound (PAINS). Here we present evidence indicating that curcumin is not a PAINS, but an inhibitor to APE1 redox function that affects many genes and pathways. This discovery explains the wide range of effects of curcumin on diverse human diseases and predicts a potential application in treatment of viral infection and virus-associated cancer. As a proof-of-concept, we demonstrated that curcumin is able to efficiently block Kaposi's sarcoma-associated herpesvirus replication and inhibit the pathogenic processes of angiogenesis and cell invasion.
Collapse
Affiliation(s)
- He Li
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA; Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - Canrong Zhong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qian Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weikang Chen
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yan Yuan
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
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 PMCID: PMC7250474 DOI: 10.1124/jpet.118.248088] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
49
|
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: 4.7] [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
|
50
|
Bhat AA, Lu H, Soutto M, Capobianco A, Rai P, Zaika A, El-Rifai W. Exposure of Barrett's and esophageal adenocarcinoma cells to bile acids activates EGFR-STAT3 signaling axis via induction of APE1. Oncogene 2018; 37:6011-6024. [PMID: 29991802 PMCID: PMC6328352 DOI: 10.1038/s41388-018-0388-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 02/06/2023]
Abstract
The development of Barret’s esophagus (BE) and its progression to esophageal adenocarcinoma (EAC) is highly linked to exposure to acidic bile salts due to chronic gastroesophageal reflux disease (GERD). In this study, we investigated the role of Apurinic/apyrimidinic endonuclease 1 /redox effector factor-1 (APE-1/REF-1) in STAT3 activation in response to EAC. Our results indicate that APE1 is constitutively overexpressed in EAC whereas its expression is transiently induced in response to acidic bile salts in non-neoplastic BE. Using overexpression or shRNA knockdown of APE1, we found that APE1 is required for phosphorylation, nuclear localization, and transcription activation of STAT3. By using an APE1 redox-specific mutant (C65A) and APE1 redox inhibitor (E3330), we demonstrate that APE1 activates STAT3 in a redox-dependent manner. By using pharmacologic inhibitors and genetic knockdown systems, we found that EGFR is a required link between APE1 and STAT3. EGFR phosphorylation (Y1068) was directly associated with APE1 levels and redox function. Co-immunoprecipitation and proximity ligation assays indicated that APE-1 coexists and interacts with the EGFR-STAT3 protein complex. Consistent with these findings, we demonstrated a significant induction in mRNA expression levels of STAT3 target genes (IL-6, IL-17A, BCL-xL, Survivin and c-Myc) in BE and EAC cells, following acidic bile salts treatment. ChIP assays indicated that acidic bile salts treatment enhances binding of STAT3 to the promoter of its target genes, Survivin and BCL-xL. Inhibition of APE1/REF-1 redox activity using E3330 abrogated STAT3 DNA binding and transcriptional activity. The induction of APE-1 - STAT3 axis in acidic bile salts conditions provided a survival advantage and promoted cellular proliferation. In summary, our study provides multiple pieces of evidence supporting a critical role for APE1 induction in activating the EGFR-STAT3 signaling axis in response to acidic bile salts, the main risk factors for Barrett’s carcinogenesis.
Collapse
Affiliation(s)
- Ajaz A Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.,Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Mohammed Soutto
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anthony Capobianco
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Priyamvada Rai
- Department of Medicine, Division of Medical Oncology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alexander Zaika
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.,Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA. .,Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA.
| |
Collapse
|