1
|
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
|
2
|
Chen T, Xiao Z, Liu X, Wang T, Wang Y, Ye F, Su J, Yao X, Xiong L, Yang DH. Natural products for combating multidrug resistance in cancer. Pharmacol Res 2024; 202:107099. [PMID: 38342327 DOI: 10.1016/j.phrs.2024.107099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Cancer cells frequently develop resistance to chemotherapeutic therapies and targeted drugs, which has been a significant challenge in cancer management. With the growing advances in technologies in isolation and identification of natural products, the potential of natural products in combating cancer multidrug resistance has received substantial attention. Importantly, natural products can impact multiple targets, which can be valuable in overcoming drug resistance from different perspectives. In the current review, we will describe the well-established mechanisms underlying multidrug resistance, and introduce natural products that could target these multidrug resistant mechanisms. Specifically, we will discuss natural compounds such as curcumin, resveratrol, baicalein, chrysin and more, and their potential roles in combating multidrug resistance. This review article aims to provide a systematic summary of recent advances of natural products in combating cancer drug resistance, and will provide rationales for novel drug discovery.
Collapse
Affiliation(s)
- Ting Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zhicheng Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xiaoyan Liu
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Tingfang Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yun Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Fei Ye
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Juan Su
- School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Xuan Yao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Liyan Xiong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, NY 11501, USA.
| |
Collapse
|
3
|
Lautrup S, Myrup Holst C, Yde A, Asmussen S, Thinggaard V, Larsen K, Laursen LS, Richner M, Vægter CB, Prieto GA, Berchtold N, Cotman CW, Stevnsner T. The role of aging and brain-derived neurotrophic factor signaling in expression of base excision repair genes in the human brain. Aging Cell 2023; 22:e13905. [PMID: 37334527 PMCID: PMC10497833 DOI: 10.1111/acel.13905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 06/20/2023] Open
Abstract
DNA damage is a central contributor to the aging process. In the brain, a major threat to the DNA is the considerable amount of reactive oxygen species produced, which can inflict oxidative DNA damage. This type of damage is removed by the base excision repair (BER) pathway, an essential DNA repair mechanism, which contributes to genome stability in the brain. Despite the crucial role of the BER pathway, insights into how this pathway is affected by aging in the human brain and the underlying regulatory mechanisms are very limited. By microarray analysis of four cortical brain regions from humans aged 20-99 years (n = 57), we show that the expression of core BER genes is largely downregulated during aging across brain regions. Moreover, we find that expression of many BER genes correlates positively with the expression of the neurotrophin brain-derived neurotrophic factor (BDNF) in the human brain. In line with this, we identify binding sites for the BDNF-activated transcription factor, cyclic-AMP response element-binding protein (CREB), in the promoter of most BER genes and confirm the ability of BDNF to regulate several BER genes by BDNF treatment of mouse primary hippocampal neurons. Together, these findings uncover the transcriptional landscape of BER genes during aging of the brain and suggest BDNF as an important regulator of BER in the human brain.
Collapse
Affiliation(s)
- Sofie Lautrup
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Department of Clinical Molecular BiologyUniversity of Oslo and Akershus University HospitalLørenskogNorway
| | | | - Anne Yde
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Stine Asmussen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Vibeke Thinggaard
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Knud Larsen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | | | - Mette Richner
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular MedicineAarhus UniversityAarhusDenmark
| | - Christian B. Vægter
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular MedicineAarhus UniversityAarhusDenmark
| | - G. Aleph Prieto
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Instituto de NeurobiologíaUNAM‐JuriquillaJuriquillaMexico
| | - Nicole Berchtold
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Carl W. Cotman
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Tinna Stevnsner
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| |
Collapse
|
4
|
Siswanto FM, Okukawa K, Tamura A, Oguro A, Imaoka S. Hydrogen peroxide activates APE1/Ref-1 via NF-κB and Parkin: A role in liver cancer resistance to oxidative stress. Free Radic Res 2023:1-31. [PMID: 37364176 DOI: 10.1080/10715762.2023.2229509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/09/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Cancer cells exhibit an altered redox balance and aberrant redox signaling due to genetic, metabolic, and microenvironment-associated reprogramming. Persistently elevated levels of reactive oxygen species (ROS) contribute to many aspects of tumor development and progression. Emerging studies demonstrated the vital role of apurinic/apyrimidinic endonuclease 1 or reduction/oxidation (redox) factor 1(APE1/Ref-1) in the oxidative stress response and survival of cancer cells. APE1/Ref-1 is a multifunctional enzyme involved in the DNA damage response and functions as a redox regulator of transcription factors. We herein demonstrated that basal hydrogen peroxide (H2O2) and APE1/Ref-1 expression levels were markedly higher in cancer cell lines than in non-cancerous cells. Elevated APE1/Ref-1 levels were associated with shorter survival in liver cancer patients. Mechanistically, we showed that H2O2 activated nuclear factor-κB (NF-κB). RelA/p65 inhibited the expression of the E3 ubiquitin ligase Parkin, possibly by interfering with ATF4 activity. Parkin was responsible for the ubiquitination and proteasomal degradation of APE1/Ref-1; therefore, the H2O2-induced suppression of Parkin expression increased APE1/Ref-1 levels. The probability of survival was lower in liver cancer patients with low Parkin and high RelA expression levels. Additionally, Parkin and RelA expression levels negatively and positively correlated with APE1/Ref-1 levels, respectively, in the TCGA liver cancer cohort. We concluded that increases in APE1/Ref-1 via the NF-κB and Parkin pathways are critical for cancer cell survival under oxidative stress. The present results show the potential of the NF-κB-Parkin-APE1/Ref-1 axis as a prognostic factor and therapeutic strategy to eradicate liver cancer.
Collapse
Affiliation(s)
- Ferbian Milas Siswanto
- Department of Biomedical Chemistry, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
- Department of Biochemistry, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Kenta Okukawa
- Department of Biomedical Chemistry, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| | - Akiyoshi Tamura
- Department of Biomedical Chemistry, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| | - Ami Oguro
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Susumu Imaoka
- Department of Biomedical Chemistry, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| |
Collapse
|
5
|
Lee EO, Joo HK, Lee YR, Kim S, Lee KH, Lee SD, Jeon BH. APE1/Ref-1 Inhibits Adipogenic Transcription Factors during Adipocyte Differentiation in 3T3-L1 Cells. Int J Mol Sci 2023; 24:ijms24043251. [PMID: 36834665 PMCID: PMC9961804 DOI: 10.3390/ijms24043251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a multifunctional protein involved in DNA repair and redox regulation. The redox activity of APE1/Ref-1 is involved in inflammatory responses and regulation of DNA binding of transcription factors related to cell survival pathways. However, the effect of APE1/Ref-1 on adipogenic transcription factor regulation remains unknown. In this study, we investigated the effect of APE1/Ref-1 on the regulation of adipocyte differentiation in 3T3-L1 cells. During adipocyte differentiation, APE1/Ref-1 expression significantly decreased with the increased expression of adipogenic transcription factors such as CCAAT/enhancer binding protein (C/EBP)-α and peroxisome proliferator-activated receptor (PPAR)-γ, and the adipocyte differentiation marker adipocyte protein 2 (aP2) in a time-dependent manner. However, APE1/Ref-1 overexpression inhibited C/EBP-α, PPAR-γ, and aP2 expression, which was upregulated during adipocyte differentiation. In contrast, silencing APE1/Ref-1 or redox inhibition of APE1/Ref-1 using E3330 increased the mRNA and protein levels of C/EBP-α, PPAR-γ, and aP2 during adipocyte differentiation. These results suggest that APE1/Ref-1 inhibits adipocyte differentiation by regulating adipogenic transcription factors, suggesting that APE1/Ref-1 is a potential therapeutic target for regulating adipocyte differentiation.
Collapse
Affiliation(s)
- Eun-Ok Lee
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
| | - Hee-Kyoung Joo
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
| | - Yu-Ran Lee
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
| | - Sungmin Kim
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
| | - Kwon-Ho Lee
- Department of Physical Therapy, Joongbu University, 201 Daehak-ro, Geumsan-gun 32713, Chungcheongnam-do, Republic of Korea
| | - Sang-Do Lee
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
| | - Byeong-Hwa Jeon
- Research Institute of Medical Sciences, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Jung-gu, Republic of Korea
- Correspondence: ; Tel.: +82-42-580-8214
| |
Collapse
|
6
|
Rios-Covian D, Butcher LD, Ablack AL, den Hartog G, Matsubara MT, Ly H, Oates AW, Xu G, Fisch KM, Ahrens ET, Toden S, Brown CC, Kim K, Le D, Eckmann L, Dhar B, Izumi T, Ernst PB, Crowe SE. A Novel Hypomorphic Apex1 Mouse Model Implicates Apurinic/Apyrimidinic Endonuclease 1 in Oxidative DNA Damage Repair in Gastric Epithelial Cells. Antioxid Redox Signal 2023; 38:183-197. [PMID: 35754343 PMCID: PMC10039277 DOI: 10.1089/ars.2021.0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/20/2023]
Abstract
Aims: Though best known for its role in oxidative DNA damage repair, apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that regulates multiple host responses during oxidative stress, including the reductive activation of transcription factors. As knockout of the APE1-encoding gene, Apex1, is embryonically lethal, we sought to create a viable model with generalized inhibition of APE1 expression. Results: A hypomorphic (HM) mouse with decreased APE1 expression throughout the body was generated using a construct containing a neomycin resistance (NeoR) cassette knocked into the Apex1 site. Offspring were assessed for APE1 expression, breeding efficiency, and morphology with a focused examination of DNA damage in the stomach. Heterozygotic breeding pairs yielded 50% fewer HM mice than predicted by Mendelian genetics. APE1 expression was reduced up to 90% in the lungs, heart, stomach, and spleen. The HM offspring were typically smaller, and most had a malformed tail. Oxidative DNA damage was increased spontaneously in the stomachs of HM mice. Further, all changes were reversed when the NeoR cassette was removed. Primary gastric epithelial cells from HM mice differentiated more quickly and had more evidence of oxidative DNA damage after stimulation with Helicobacter pylori or a chemical carcinogen than control lines from wildtype mice. Innovation: A HM mouse with decreased APE1 expression throughout the body was generated and extensively characterized. Conclusion: The results suggest that HM mice enable studies of APE1's multiple functions throughout the body. The detailed characterization of the stomach showed that gastric epithelial cells from HM were more susceptible to DNA damage. Antioxid. Redox Signal. 38, 183-197.
Collapse
Affiliation(s)
- David Rios-Covian
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Lindsay D. Butcher
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Amber L. Ablack
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Gerco den Hartog
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Mason T. Matsubara
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Hong Ly
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Andrew W. Oates
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Guorong Xu
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Kathleen M. Fisch
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Eric T. Ahrens
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Shusuke Toden
- Molecular Stethoscope, Inc., San Diego, California, USA
| | - Corrie C. Brown
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Kenneth Kim
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Dzung Le
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Lars Eckmann
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Bithika Dhar
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Tadahide Izumi
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Peter B. Ernst
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Immunology, Chiba University, Chiba, Japan
| | - Sheila E. Crowe
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
7
|
Zhao J, Huai J. Role of primary aging hallmarks in Alzheimer´s disease. Theranostics 2023; 13:197-230. [PMID: 36593969 PMCID: PMC9800733 DOI: 10.7150/thno.79535] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which severely threatens the health of the elderly and causes significant economic and social burdens. The causes of AD are complex and include heritable but mostly aging-related factors. The primary aging hallmarks include genomic instability, telomere wear, epigenetic changes, and loss of protein stability, which play a dominant role in the aging process. Although AD is closely associated with the aging process, the underlying mechanisms involved in AD pathogenesis have not been well characterized. This review summarizes the available literature about primary aging hallmarks and their roles in AD pathogenesis. By analyzing published literature, we attempted to uncover the possible mechanisms of aberrant epigenetic markers with related enzymes, transcription factors, and loss of proteostasis in AD. In particular, the importance of oxidative stress-induced DNA methylation and DNA methylation-directed histone modifications and proteostasis are highlighted. A molecular network of gene regulatory elements that undergoes a dynamic change with age may underlie age-dependent AD pathogenesis, and can be used as a new drug target to treat AD.
Collapse
|
8
|
Xue Z, Demple B. Knockout and Inhibition of Ape1: Roles of Ape1 in Base Excision DNA Repair and Modulation of Gene Expression. Antioxidants (Basel) 2022; 11:antiox11091817. [PMID: 36139891 PMCID: PMC9495735 DOI: 10.3390/antiox11091817] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Apurinic/apyrimidinic endonuclease 1/redox effector-1 (Ape1/Ref-1) is the major apurinic/apyrimidinic (AP) endonuclease in mammalian cells. It functions mainly in the base excision repair pathway to create a suitable substrate for DNA polymerases. Human Ape1 protein can activate some transcription factors to varying degrees, dependent on its N-terminal, unstructured domain, and some of the cysteines within it, apparently via a redox mechanism in some cases. Many cancer studies also suggest that Ape1 has potential for prognosis in terms of the protein level or intracellular localization. While homozygous disruption of the Ape1 structural gene APEX1 in mice causes embryonic lethality, and most studies in cell culture indicate that the expression of Ape1 is essential, some recent studies reported the isolation of viable APEX1 knockout cells with only mild phenotypes. It has not been established by what mechanism the Ape1-null cell lines cope with the endogenous DNA damage that the enzyme normally handles. We review the enzymatic and other activities of Ape1 and the recent studies of the properties of the APEX1 knockout lines. The APEX1 deletions in CH12F3 and HEK293 FT provide an opportunity to test for possible off-target effects of Ape1 inhibition. For this work, we tested the Ape1 endonuclease inhibitor Compound 3 and the redox inhibitor APX2009. Our results confirmed that both APEX1 knockout cell lines are modestly more sensitive to killing by an alkylating agent than their Ape1-proficient cells. Surprisingly, the knockout lines showed equal sensitivity to direct killing by either inhibitor, despite the lack of the target protein. Moreover, the CH12F3 APEX1 knockout was even more sensitive to Compound 3 than its APEX1+ counterpart. Thus, it appears that both Compound 3 and APX2009 have off-target effects. In cases where this issue may be important, it is advisable that more specific endpoints than cell survival be tested for establishing mechanism.
Collapse
Affiliation(s)
- Zhouyiyuan Xue
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA
- Molecular and Cellular Biochemistry Program, Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Bruce Demple
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA
- Correspondence: ; Tel.: +1-(631)-444-3978
| |
Collapse
|
9
|
Excessive Folic Acid Mimics Folate Deficiency in Human Lymphocytes. Curr Issues Mol Biol 2022; 44:1452-1462. [PMID: 35723355 PMCID: PMC9164024 DOI: 10.3390/cimb44040097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/20/2022] [Indexed: 12/19/2022] Open
Abstract
Food fortification with synthetic folic acid (FA), along with supplementation, results in a marked increase in the population total of serum folates and unmetabolized folic acid (UMFA). Despite the success in reducing neural tube defects at birth in the intended target population (women of childbearing age), the potential deleterious effects of chronically high levels of UMFA in susceptible segments of the population require further investigation. In this study, we examine the effects of FA concentrations, ranging from depletion to supraphysiological levels, on markers of proliferation, DNA methylation, and DNA damage and repair in a human lymphoblastoid cell line (LCL). We note that both low and high levels of FA similarly impact global DNA methylation, cytome biomarkers measured through the CBMN assay, DNA damage induced by oxidative stress, and DNA base excision repair gene expression.
Collapse
|
10
|
The Timing and Duration of Folate Restriction Differentially Impacts Colon Carcinogenesis. Nutrients 2021; 14:nu14010016. [PMID: 35010891 PMCID: PMC8746403 DOI: 10.3390/nu14010016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Diet plays a crucial role in the development of colorectal cancer (CRC). Of particular importance, folate, present in foods and supplements, is a crucial modulator of CRC risk. The role of folate, and, specifically, the synthetic variant, folic acid, in the primary prevention of CRC has not been fully elucidated. Animal studies varied considerably in the timing, duration, and supplementation of folates, leading to equivocal results. Our work attempts to isolate these variables to ascertain the role of folic acid in CRC initiation, as we previously demonstrated that folate restriction conferred protection against CRC initiation in a β-pol haploinsufficient mouse model. Here we demonstrated that prior adaptation to folate restriction altered the response to carcinogen exposure in wild-type C57BL/6 mice. Mice adapted to folate restriction for 8 weeks were protected from CRC initiation compared to mice placed on folate restriction for 1 week, irrespective of antibiotic supplementation. Through analyses of mTOR signaling, DNA methyltransferase, and DNA repair, we have identified factors that may play a critical role in the differential responses to folate restriction. Furthermore, the timing and duration of folate restriction altered these pathways differently in the absence of carcinogenic insult. These results represent novel findings, as we were able to show that, in the same model and under controlled conditions, folate restriction produced contrasting results depending on the timing and duration of the intervention.
Collapse
|
11
|
El-Shetry ES, Mohamed AAR, Khater SI, Metwally MMM, Nassan MA, Shalaby S, A M El-Mandrawy S, Bin Emran T, M Abdel-Ghany H. Synergistically enhanced apoptotic and oxidative DNA damaging pathways in the rat brain with lead and/or aluminum metals toxicity: Expression pattern of genes OGG1 and P53. J Trace Elem Med Biol 2021; 68:126860. [PMID: 34583094 DOI: 10.1016/j.jtemb.2021.126860] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Lead (Pb) and aluminum (Al) are ubiquitous environmental pollutants and are known to induce neurodegenerative disorders. They enhance neuronal changes and may involve glial alterations and other consequences. We intend to evaluate the mechanism through which the long-term exposure to Pb acetate alone or in combination with aluminum-chloride induced neurological impacts in rats. METHODS For this aim, a total number of forty male Sprague Dawley rats were assigned into four groups. Control (DW), Pb acetate (12.5 mg/kg BW), Al chloride (64 mg/kg BW), and the combination group were experimentally exposed for 60 days. Biochemical evaluation of oxidative stress biomarkers, transcriptional-mediated changes in the expression pattern of OGG1 and P53 genes by qRT-PCR were applied. Histopathological modifications in the brain tissue with immunohistochemical reactivity of GFAP were also detected. RESULTS Our findings revealed that lipid peroxidation was markedly enhanced but inhibited antioxidant enzyme activity in brain tissue in all exposed groups regarding the control. Pb-acetate elevated the biochemical concentration of dopamine and serotonin while AlCl3 declined their levels in the brain homogenate of rats. Furthermore, the exposure to one or both metals elevated the comet assay indices and serum level of 8-hydroxy-2' -deoxyguanosine, up-regulated the expression of P53, OGG1 and GFAP immunoreactivity in the central nervous system. Histologically, they caused several brain tissue alterations. CONCLUSION The exposure to Pb and/or Al could be key candidates for neurodegenerative changes in the brain of rats via oxidative, apoptotic, and DNA damaging pathways. Besides, according to our findings, exposure to both Pb acetate and Aluminium chloride have synergistic damaging effects on the central nervous system of rats. Also, they have opposing effects on the secretion of monoamine neurotransmitters DA and 5 H T.
Collapse
Affiliation(s)
- Eman S El-Shetry
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Amany Abdel-Rahman Mohamed
- Departments of Forensic Medicine and Toxicology and Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| | - Safaa I Khater
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, 4511, Egypt
| | - Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mohamed A Nassan
- Department of clinical laboratory sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shimaa Shalaby
- Department of Physiology, Faculty of Vet. Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Shefaa A M El-Mandrawy
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Heba M Abdel-Ghany
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
12
|
Fleming AM, Burrows CJ. Oxidative stress-mediated epigenetic regulation by G-quadruplexes. NAR Cancer 2021; 3:zcab038. [PMID: 34541539 PMCID: PMC8445369 DOI: 10.1093/narcan/zcab038] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Many cancer-associated genes are regulated by guanine (G)-rich sequences that are capable of refolding from the canonical duplex structure to an intrastrand G-quadruplex. These same sequences are sensitive to oxidative damage that is repaired by the base excision repair glycosylases OGG1 and NEIL1–3. We describe studies indicating that oxidation of a guanosine base in a gene promoter G-quadruplex can lead to up- and downregulation of gene expression that is location dependent and involves the base excision repair pathway in which the first intermediate, an apurinic (AP) site, plays a key role mediated by AP endonuclease 1 (APE1/REF1). The nuclease activity of APE1 is paused at a G-quadruplex, while the REF1 capacity of this protein engages activating transcription factors such as HIF-1α, AP-1 and p53. The mechanism has been probed by in vitro biophysical studies, whole-genome approaches and reporter plasmids in cellulo. Replacement of promoter elements by a G-quadruplex sequence usually led to upregulation, but depending on the strand and precise location, examples of downregulation were also found. The impact of oxidative stress-mediated lesions in the G-rich sequence enhanced the effect, whether it was positive or negative.
Collapse
Affiliation(s)
- Aaron M Fleming
- Department of Chemistry, University of Utah, 315 S. 1400 East, Salt Lake City, UT 84112-0850, USA
| | - Cynthia J Burrows
- Department of Chemistry, University of Utah, 315 S. 1400 East, Salt Lake City, UT 84112-0850, USA
| |
Collapse
|
13
|
Davletgildeeva AT, Ishchenko AA, Saparbaev M, Fedorova OS, Kuznetsov NA. The Enigma of Substrate Recognition and Catalytic Efficiency of APE1-Like Enzymes. Front Cell Dev Biol 2021; 9:617161. [PMID: 33842455 PMCID: PMC8033172 DOI: 10.3389/fcell.2021.617161] [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] [Received: 10/14/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Despite significant achievements in the elucidation of the nature of protein-DNA contacts that control the specificity of nucleotide incision repair (NIR) by apurinic/apyrimidinic (AP) endonucleases, the question on how a given nucleotide is accommodated by the active site of the enzyme remains unanswered. Therefore, the main purpose of our study was to compare kinetics of conformational changes of three homologous APE1-like endonucleases (insect Drosophila melanogaster Rrp1, amphibian Xenopus laevis xAPE1, and fish Danio rerio zAPE1) during their interaction with various damaged DNA substrates, i.e., DNA containing an F-site (an uncleavable by DNA-glycosylases analog of an AP-site), 1,N6-ethenoadenosine (εA), 5,6-dihydrouridine (DHU), uridine (U), or the α-anomer of adenosine (αA). Pre-steady-state analysis of fluorescence time courses obtained for the interaction of the APE1-like enzymes with DNA substrates containing various lesions allowed us to outline a model of substrate recognition by this class of enzymes. It was found that the differences in rates of DNA substrates’ binding do not lead to significant differences in the cleavage efficiency of DNA containing a damaged base. The results suggest that the formation of enzyme–substrate complexes is not the key factor that limits enzyme turnover; the mechanisms of damage recognition and cleavage efficacy are related to fine conformational tuning inside the active site.
Collapse
Affiliation(s)
- Anastasiia T Davletgildeeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Alexander A Ishchenko
- Group "Mechanisms of DNA Repair and Carcinogenesis", Equipe Labellisée LIGUE 2016, CNRS UMR 9019, Université Paris-Saclay, Villejuif, France
| | - Murat Saparbaev
- Group "Mechanisms of DNA Repair and Carcinogenesis", Equipe Labellisée LIGUE 2016, CNRS UMR 9019, Université Paris-Saclay, Villejuif, France
| | - Olga S Fedorova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nikita A Kuznetsov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
14
|
Yu Y, Li L, Li G, Zhou X, Deng T, Liang M, Nie G. Intracellular enzyme-powered DNA circuit with a tunable amplifier for miRNA imaging. Chem Commun (Camb) 2021; 57:3753-3756. [PMID: 33876121 DOI: 10.1039/d1cc00536g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We describe an intracellular enzyme-powered DNA circuit probe with a tunable amplifier for sensitive and selective detection of miRNA. This approach has been successfully applied for in situ miRNA-21 fluorescence imaging in live cells. Also, we used chemicals to elevate the APE1 expression level rendering a tunable amplification strength for more flexible imaging applications.
Collapse
Affiliation(s)
- Yingjie Yu
- Institute of Translational Medicine, Department of Otolaryngology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen 518035, China.
| | | | | | | | | | | | | |
Collapse
|
15
|
Kladova OA, Iakovlev DA, Groisman R, Ishchenko AA, Saparbaev MK, Fedorova OS, Kuznetsov NA. An Assay for the Activity of Base Excision Repair Enzymes in Cellular Extracts Using Fluorescent DNA Probes. BIOCHEMISTRY (MOSCOW) 2021; 85:480-489. [PMID: 32569555 DOI: 10.1134/s0006297920040082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Damaged DNA bases are removed by the base excision repair (BER) mechanism. This enzymatic process begins with the action of one of DNA glycosylases, which recognize damaged DNA bases and remove them by hydrolyzing N-glycosidic bonds with the formation of apurinic/apyrimidinic (AP) sites. Apurinic/apyrimidinic endonuclease 1 (APE1) hydrolyzes the phosphodiester bond on the 5'-side of the AP site with generation of the single-strand DNA break. A decrease in the functional activity of BER enzymes is associated with the increased risk of cardiovascular, neurodegenerative, and oncological diseases. In this work, we developed a fluorescence method for measuring the activity of key human DNA glycosylases and AP endonuclease in cell extracts. The efficacy of fluorescent DNA probes was tested using purified enzymes; the most efficient probes were tested in the enzymatic activity assays in the extracts of A549, MCF7, HeLa, WT-7, HEK293T, and HKC8 cells. The activity of enzymes responsible for the repair of AP sites and removal of uracil and 5,6-dihydrouracil residues was higher in cancer cell lines as compared to the normal HKC8 human kidney cell line.
Collapse
Affiliation(s)
- O A Kladova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - D A Iakovlev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - R Groisman
- Groupe "Réparation de l'AND", Equipe Labellisée par la Ligue Nationale contre le Cancer, CNRS UMR 8200, Université Paris-Sud, Université Paris-Saclay, Villejuif, F-94805, France.,Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - A A Ishchenko
- Groupe "Réparation de l'AND", Equipe Labellisée par la Ligue Nationale contre le Cancer, CNRS UMR 8200, Université Paris-Sud, Université Paris-Saclay, Villejuif, F-94805, France.,Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - M K Saparbaev
- Groupe "Réparation de l'AND", Equipe Labellisée par la Ligue Nationale contre le Cancer, CNRS UMR 8200, Université Paris-Sud, Université Paris-Saclay, Villejuif, F-94805, France.,Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - O S Fedorova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia
| | - N A Kuznetsov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia
| |
Collapse
|
16
|
The role of active-site amino acid residues in the cleavage of DNA and RNA substrates by human apurinic/apyrimidinic endonuclease APE1. Biochim Biophys Acta Gen Subj 2020; 1864:129718. [PMID: 32858086 DOI: 10.1016/j.bbagen.2020.129718] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Human apurinic/apyrimidinic endonuclease APE1 is one of participants of the DNA base excision repair pathway. APE1 processes AP-sites and many other types of DNA damage via hydrolysis of the phosphodiester bond on the 5' side of the lesion. APE1 also acts as an endoribonuclease, i.e., can cleave undamaged RNA. METHODS Using pre-steady-state kinetic analysis we examined the role of certain catalytically important amino acids in APE1 enzymatic pathway and described their involvement in the mechanism of the target nucleotide recognition. RESULTS Comparative analysis of the cleavage efficiency of damaged DNAs containing an abasic site, 5,6-dihydrouridine, or α-anomer of adenosine as well as 3'-5'-exonuclease degradation of undamaged DNA and endonuclease hydrolysis of RNA substrates by mutant APE1 enzymes containing a substitution of an active-site amino acid residue (D210N, N212A, T268D, M270A, or D308A) was performed. Detailed pre-steady-state kinetics of conformational changes of the enzyme and of DNA substrate molecules during recognition and cleavage of the abasic site were studied. CONCLUSIONS It was revealed that substitution T268D significantly disturbed initial DNA binding, whereas Asn212 is critical for the DNA-bending stage and catalysis. Substitution D210N increased the binding efficacy and blocked the catalytic reaction, but D308A decreased the binding efficacy owing to disruption of Mg2+ coordination. Finally, the substitution of Met270 also destabilized the enzyme-substrate complex but did not affect the catalytic reaction. SIGNIFICANCE It was found that the tested substitutions of the active-site amino acid residues affected different stages of the complex formation process as well as the catalytic reaction.
Collapse
|
17
|
Kim DV, Makarova AV, Miftakhova RR, Zharkov DO. Base Excision DNA Repair Deficient Cells: From Disease Models to Genotoxicity Sensors. Curr Pharm Des 2020; 25:298-312. [PMID: 31198112 DOI: 10.2174/1381612825666190319112930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 12/29/2022]
Abstract
Base excision DNA repair (BER) is a vitally important pathway that protects the cell genome from many kinds of DNA damage, including oxidation, deamination, and hydrolysis. It involves several tightly coordinated steps, starting from damaged base excision and followed by nicking one DNA strand, incorporating an undamaged nucleotide, and DNA ligation. Deficiencies in BER are often embryonic lethal or cause morbid diseases such as cancer, neurodegeneration, or severe immune pathologies. Starting from the early 1980s, when the first mammalian cell lines lacking BER were produced by spontaneous mutagenesis, such lines have become a treasure trove of valuable information about the mechanisms of BER, often revealing unexpected connections with other cellular processes, such as antibody maturation or epigenetic demethylation. In addition, these cell lines have found an increasing use in genotoxicity testing, where they provide increased sensitivity and representativity to cell-based assay panels. In this review, we outline current knowledge about BER-deficient cell lines and their use.
Collapse
Affiliation(s)
- Daria V Kim
- Novosibirsk State University, 2 Pirogova St., Novosibirsk 630090, Russian Federation
| | - Alena V Makarova
- RAS Institute of Molecular Genetics, 2 Kurchatova Sq., Moscow 123182, Russian Federation
| | - Regina R Miftakhova
- Kazan Federal University, 18 Kremlevsakaya St., Kazan 420008, Russian Federation
| | - Dmitry O Zharkov
- Novosibirsk State University, 2 Pirogova St., Novosibirsk 630090, Russian Federation.,SB RAS Institute of Chemical Biology and Fu ndamental Medicine, 8 Lavrentieva Ave., Novosibirsk 630090, Russian Federation
| |
Collapse
|
18
|
Alekseeva IV, Bakman AS, Vorobjev YN, Fedorova OS, Kuznetsov NA. Role of Ionizing Amino Acid Residues in the Process of DNA Binding by Human AP Endonuclease 1 and in Its Catalysis. J Phys Chem B 2019; 123:9546-9556. [PMID: 31633353 DOI: 10.1021/acs.jpcb.9b07150] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the repair of the damage to bases, human apurinic/apyrimidinic (AP) endonuclease 1 (APE1) is a key participant via the DNA base excision repair pathway. APE1 cleaves AP sites in DNA, which are potentially cytotoxic and highly mutagenic if left unrepaired. According to existing structural data, this enzyme's active site contains many polar amino acid residues, which form extensive contacts with a DNA substrate. A few alternative catalytic mechanisms of the phosphodiester bond hydrolysis by APE1 have been reported. Here, the kinetics of conformational changes of the enzyme and of DNA substrate molecules were studied during the recognition and cleavage of the abasic site in the pH range from 5.5 to 9.0 using stopped-flow fluorescence techniques. The activity of APE1 increased with an increase in pH because of acceleration of the rates of catalytic complex formation and of the catalytic reaction. Molecular dynamics simulation uncovered a significant increase in the pKa of His-309 located in the active site of the enzyme. This finding revealed that the observed enhancement of enzymatic activity with pH could be associated with deprotonation of not only Tyr-171 but also His-309. The obtained data allowed us to hypothesize that the ionized state of these residues could be a molecular switch between the alternative catalytic mechanisms, which involve different functionalities of these residues throughout the reaction.
Collapse
Affiliation(s)
- Irina V Alekseeva
- Institute of Chemical Biology and Fundamental Medicine , Novosibirsk 630090 , Russia
| | - Artemiy S Bakman
- Institute of Chemical Biology and Fundamental Medicine , Novosibirsk 630090 , Russia
| | - Yury N Vorobjev
- Institute of Chemical Biology and Fundamental Medicine , Novosibirsk 630090 , Russia
| | - Olga S Fedorova
- Institute of Chemical Biology and Fundamental Medicine , Novosibirsk 630090 , Russia.,Department of Natural Sciences , Novosibirsk State University , Novosibirsk 630090 , Russia
| | - Nikita A Kuznetsov
- Institute of Chemical Biology and Fundamental Medicine , Novosibirsk 630090 , Russia.,Department of Natural Sciences , Novosibirsk State University , Novosibirsk 630090 , Russia
| |
Collapse
|
19
|
Li M, Yang X, Lu X, Dai N, Zhang S, Cheng Y, Zhang L, Yang Y, Liu Y, Yang Z, Wang D, Wilson DM. APE1 deficiency promotes cellular senescence and premature aging features. Nucleic Acids Res 2019; 46:5664-5677. [PMID: 29750271 PMCID: PMC6009672 DOI: 10.1093/nar/gky326] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/23/2018] [Indexed: 12/27/2022] Open
Abstract
Base excision repair (BER) handles many forms of endogenous DNA damage, and apurinic/apyrimidinic endonuclease 1 (APE1) is central to this process. Deletion of both alleles of APE1 (a.k.a. Apex1) in mice leads to embryonic lethality, and deficiency in cells can promote cell death. Unlike most other BER proteins, APE1 expression is inversely correlated with cellular senescence in primary human fibroblasts. Depletion of APE1 via shRNA induced senescence in normal human BJ fibroblasts, a phenotype that was not seen in counterpart cells expressing telomerase. APE1 knock-down in primary fibroblasts resulted in global DNA damage accumulation, and the induction of p16INK4a and p21WAF1 stress response pathways; the DNA damage response, as assessed by γ-H2AX, was particularly pronounced at telomeres. Conditional knock-out of Apex1 in mice at post-natal day 7/12 resulted in impaired growth, reduced organ size, and increased cellular senescence. The effect of Apex1 deletion at post-natal week 6 was less obvious, other than cellular senescence, until ∼8-months of age, when premature aging characteristics, such as hair loss and impaired wound healing, were seen. Low APE1 expression in patient cancer tissue also correlated with increased senescence. Our results point to a key role for APE1 in regulating cellular senescence and aging features, with telomere status apparently affecting the outcome.
Collapse
Affiliation(s)
- Mengxia Li
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Xiao Yang
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Xianfeng Lu
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Nan Dai
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Shiheng Zhang
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Yi Cheng
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Lei Zhang
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Yuxin Yang
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Yie Liu
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Ste. 100, Baltimore, MD 21224, USA
| | - Zhenzhou Yang
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - Dong Wang
- Cancer Center of Daping Hospital, Third Military Medical University, No. 10 Changjiang Zhi Rd., Yuzhong Dist., Chongqing 400042 China
| | - David M Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Ste. 100, Baltimore, MD 21224, USA
| |
Collapse
|
20
|
Alekseeva IV, Davletgildeeva AT, Arkova OV, Kuznetsov NA, Fedorova OS. The impact of single-nucleotide polymorphisms of human apurinic/apyrimidinic endonuclease 1 on specific DNA binding and catalysis. Biochimie 2019; 163:73-83. [PMID: 31150756 DOI: 10.1016/j.biochi.2019.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
Abstract
Human apurinic/apyrimidinic (AP) endonuclease APE1 is a crucial enzyme of the base excision repair (BER) pathway, which is in charge of recognition and initiation of removal of AP-sites in DNA. It is known that some single-nucleotide polymorphism (SNP) variants of APE1 have a reduced activity as compared to wild-type APE1. It has been hypothesized that genetic variation in APE1 might be responsible for an increased risk of some types of cancer. In the present work, analysis of SNPs of the APE1 gene was performed to select the set of variants having substitutions of amino acid residues on the surface of the enzyme globule and in the DNA-binding site, thereby affecting protein-protein interactions or the catalytic reaction, respectively. For seven APE1 variants (R221C, N222H, R237A, G241R, M270T, R274Q, and P311S), conformational dynamics and catalytic activities were examined. The conformational changes in the molecules of APE1 variants and in a DNA substrate were recorded as fluorescence changes of Trp and 2-aminopurine residues, respectively, using the stopped-flow technique. The results made it possible to determine the kinetic mechanism underlying the interactions of the APE1 variants with DNA substrates, to calculate the rate constants of the elementary stages, and to identify the stages of the process affected by mutation.
Collapse
Affiliation(s)
- Irina V Alekseeva
- Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentyev Ave., Novosibirsk, 630090, Russia
| | - Anastasiia T Davletgildeeva
- Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentyev Ave., Novosibirsk, 630090, Russia; Department of Natural Sciences, Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia
| | - Olga V Arkova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Lavrentyev Ave., Novosibirsk, 630090, Russia
| | - Nikita A Kuznetsov
- Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentyev Ave., Novosibirsk, 630090, Russia; Department of Natural Sciences, Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia.
| | - Olga S Fedorova
- Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentyev Ave., Novosibirsk, 630090, Russia; Department of Natural Sciences, Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia.
| |
Collapse
|
21
|
Sengar GS, Deb R, Singh U, Junghare V, Hazra S, Raja TV, Alex R, Kumar A, Alyethodi RR, Kant R, Jakshara S, Joshi CG. Identification of differentially expressed microRNAs in Sahiwal (Bos indicus) breed of cattle during thermal stress. Cell Stress Chaperones 2018; 23:1019-1032. [PMID: 29777484 PMCID: PMC6111087 DOI: 10.1007/s12192-018-0911-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 10/16/2022] Open
Abstract
microRNAs (miRNAs) are a class of small non-coding RNAs that play key roles in post transcriptional gene regulation that influence various fundamental cellular processes, including the cellular responses during environmental stresses. However, perusal of literatures revealed few reports on the differential expression of miRNA during thermal stress in Indian native (Bos indicus) cattle breeds. The present investigation aimed to identify differentially expressed miRNAs during thermal stress in Sahiwal (Bos indicus) dairy cattle breed of India, adapted with tropical climate over a long period of time. Stress responses of the animals were characterized by determining various physiological as well as biochemical parameters and differential expression profile of major heat shock protein genes. Ion Torrent deep sequencing and CLC-genomic analysis identified a set of differentially expressed miRNAs during summer and winter seasons. Most of the identified differentially expressed miRNAs were found to target heat shock responsive genes especially members of heat shock protein (HSP) family. Real-time quantification-based analysis of selected miRNAs revealed that bta-mir-1248, bta-mir-2332, bta-mir-2478, and bta-mir-1839 were significantly (p < 0.01) over expressed while bta-mir-16a, bta-let-7b, bta-mir-142, and bta-mir-425 were significantly (p < 0.01) under expressed during summer in comparison to winter. The present study enlists differentially expressed miRNAs at different environmental temperatures in Sahiwal (Bos indicus) that may be importance for further understanding the role of miRNAs on thermo-regulatory mechanisms.
Collapse
Affiliation(s)
- Gyanendra Singh Sengar
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India
- Sam Higginbottom University of Agriculture Technology & Science, Allahabad, India
| | - Rajib Deb
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India.
| | - Umesh Singh
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India
| | - Vivek Junghare
- Department of Biotechnology, Center of Nanotechnology, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - Saugata Hazra
- Department of Biotechnology, Center of Nanotechnology, Indian Institute of Technology, Roorkee, Uttarakhand, India
- Center of Nanotechnology, Indian Institute of Technology, Roorkee, Uttarakhand, India
| | - T V Raja
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India
| | - Rani Alex
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India
| | - Ashish Kumar
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India
| | - R R Alyethodi
- Molecular Genetics Laboratory, ICAR-Central Institute for Research on Cattle, -250 001, Meerut, Uttar Pradesh, India
| | - Rajiv Kant
- Sam Higginbottom University of Agriculture Technology & Science, Allahabad, India
| | - Subhash Jakshara
- Ome Research Laboratory, Anand Agricultural University, Anand, Gujarat, India
| | - C G Joshi
- Ome Research Laboratory, Anand Agricultural University, Anand, Gujarat, India
| |
Collapse
|
22
|
Fletcher SC, Grou CP, Legrand AJ, Chen X, Soderstrom K, Poletto M, Dianov GL. Sp1 phosphorylation by ATM downregulates BER and promotes cell elimination in response to persistent DNA damage. Nucleic Acids Res 2018; 46:1834-1846. [PMID: 29294106 PMCID: PMC5829641 DOI: 10.1093/nar/gkx1291] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 11/15/2022] Open
Abstract
ATM (ataxia-telangiectasia mutated) is a central molecule for DNA quality control. Its activation by DNA damage promotes cell-cycle delay, which facilitates DNA repair prior to replication. On the other hand, persistent DNA damage has been implicated in ATM-dependent cell death via apoptosis; however, the mechanisms underlying this process remain elusive. Here we find that, in response to persistent DNA strand breaks, ATM phosphorylates transcription factor Sp1 and initiates its degradation. We show that Sp1 controls expression of the key base excision repair gene XRCC1, essential for DNA strand break repair. Therefore, degradation of Sp1 leads to a vicious cycle that involves suppression of DNA repair and further aggravation of the load of DNA damage. This activates transcription of pro-apoptotic genes and renders cells susceptible to elimination via both apoptosis and natural killer cells. These findings constitute a previously unrecognized 'gatekeeper' function of ATM as a detector of cells with persistent DNA damage.
Collapse
Affiliation(s)
- Sally C Fletcher
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Claudia P Grou
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Arnaud J Legrand
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Xin Chen
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
- Department of Marine Technology, College of Ocean, Nantong University, Nantong, Jiangsu, 226007, China
| | - Kalle Soderstrom
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK
| | - Mattia Poletto
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Grigory L Dianov
- Department of Oncology, CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
- Institute of Cytology and Genetics, Russian Academy of Sciences, Lavrentyeva 10 Novosibirsk 630090, Russian Federation
- Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russian Federation
| |
Collapse
|
23
|
Hou X, Snarski P, Higashi Y, Yoshida T, Jurkevich A, Delafontaine P, Sukhanov S. Nuclear complex of glyceraldehyde-3-phosphate dehydrogenase and DNA repair enzyme apurinic/apyrimidinic endonuclease I protect smooth muscle cells against oxidant-induced cell death. FASEB J 2017; 31:3179-3192. [PMID: 28404743 DOI: 10.1096/fj.201601082r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/27/2017] [Indexed: 01/18/2023]
Abstract
Atherosclerotic plaque destabilization is the major determinant of most acute coronary events. Smooth muscle cell (SMC) death contributes to plaque destabilization. Here, we describe a novel antiapoptotic mechanism in vascular SMCs that involves interaction of nuclear glyceraldehyde-3-phosphate dehydrogenase (GAPDH) with apurinic/apyrimidinic endonuclease 1 (Ape1), the major oxidized DNA repair enzyme. GAPDH down-regulation potentiated H2O2-induced DNA damage and SMC apoptosis. Conversely, GAPDH overexpression decreased DNA damage and protected SMCs against apoptosis. Ape1 down-regulation reversed the resistance of GAPDH-overexpressing cells to DNA damage and apoptosis, which indicated that Ape1 is indispensable for GAPDH-dependent protective effects. GAPDH bound Ape1 in the SMC nucleus, and blocking (or oxidation) of GAPDH active site cysteines suppressed GAPDH/Ape1 interaction and potentiated apoptosis. GAPDH up-regulated Ape1 via a transcription factor homeobox protein Hox-A5-dependent mechanism. GAPDH levels were reduced in atherosclerotic plaque SMCs, and this effect correlated with oxidative stress and SMC apoptosis. Thus, we demonstrated that nuclear GAPDH/Ape1 interaction preserved Ape1 activity, reduced DNA damage, and prevented SMC apoptosis. Suppression of SMC apoptosis by maintenance of nuclear GAPDH/Ape1 interactions may be a novel therapy to increase atherosclerotic plaque stability.-Hou, X., Snarski, P., Higashi, Y., Yoshida, T., Jurkevich, A., Delafontaine, P., Sukhanov, S. Nuclear complex of glyceraldehyde-3-phosphate dehydrogenase and DNA repair enzyme apurinic/apyrimidinic endonuclease I protect smooth muscle cells against oxidant-induced cell death.
Collapse
Affiliation(s)
- Xuwei Hou
- Department of Medicine, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA
| | - Patricia Snarski
- Department of Medicine, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA
| | - Yusuke Higashi
- Department of Medicine, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA.,Department of Physiology and Medical Pharmacology, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA
| | - Tadashi Yoshida
- Department of Medicine, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA.,Department of Physiology and Medical Pharmacology, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA
| | - Alexander Jurkevich
- Molecular Cytology Core, University of Missouri at Columbia, Columbia, Missouri, USA
| | - Patrick Delafontaine
- Department of Medicine, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA.,Department of Physiology and Medical Pharmacology, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA
| | - Sergiy Sukhanov
- Department of Medicine, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA; .,Department of Physiology and Medical Pharmacology, School of Medicine, University of Missouri at Columbia, Columbia, Missouri, USA
| |
Collapse
|
24
|
Whitaker AM, Schaich MA, Smith MR, Flynn TS, Freudenthal BD. Base excision repair of oxidative DNA damage: from mechanism to disease. Front Biosci (Landmark Ed) 2017; 22:1493-1522. [PMID: 28199214 DOI: 10.2741/4555] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species continuously assault the structure of DNA resulting in oxidation and fragmentation of the nucleobases. Both oxidative DNA damage itself and its repair mediate the progression of many prevalent human maladies. The major pathway tasked with removal of oxidative DNA damage, and hence maintaining genomic integrity, is base excision repair (BER). The aphorism that structure often dictates function has proven true, as numerous recent structural biology studies have aided in clarifying the molecular mechanisms used by key BER enzymes during the repair of damaged DNA. This review focuses on the mechanistic details of the individual BER enzymes and the association of these enzymes during the development and progression of human diseases, including cancer and neurological diseases. Expanding on these structural and biochemical studies to further clarify still elusive BER mechanisms, and focusing our efforts toward gaining an improved appreciation of how these enzymes form co-complexes to facilitate DNA repair is a crucial next step toward understanding how BER contributes to human maladies and how it can be manipulated to alter patient outcomes.
Collapse
Affiliation(s)
- Amy M Whitaker
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, 66160
| | - Matthew A Schaich
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, 66160
| | - Mallory R Smith
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, 66160
| | - Tony S Flynn
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, 66160
| | - Bret D Freudenthal
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, 66160,
| |
Collapse
|
25
|
Illuzzi JL, McNeill DR, Bastian P, Brenerman B, Wersto R, Russell HR, Bunz F, McKinnon PJ, Becker KG, Wilson DM. Tumor-associated APE1 variant exhibits reduced complementation efficiency but does not promote cancer cell phenotypes. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:84-98. [PMID: 28181292 PMCID: PMC5321783 DOI: 10.1002/em.22074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/10/2017] [Indexed: 06/06/2023]
Abstract
Base excision repair (BER) is the major pathway for coping with most forms of endogenous DNA damage, and defects in the process have been associated with carcinogenesis. Apurinic/apyrimidinic endonuclease 1 (APE1) is a central participant in BER, functioning as a critical endonuclease in the processing of noncoding abasic sites in DNA. Evidence has suggested that APE1 missense mutants, as well as altered expression or localization of the protein, can contribute to disease manifestation. We report herein that the tumor-associated APE1 variant, R237C, shows reduced complementation efficiency of the methyl methanesulfonate hypersensitivity and impaired cell growth exhibited by APE1-deficient mouse embryonic fibroblasts. Overexpression of wild-type APE1 or the R237C variant in the nontransformed C127I mouse cell line had no effect on proliferation, cell cycle status, steady-state DNA damage levels, mitochondrial function, or cellular transformation. A human cell line heterozygous for an APE1 knockout allele had lower levels of endogenous APE1, increased cellular sensitivity to DNA-damaging agents, impaired proliferation with time, and a distinct global gene expression pattern consistent with a stress phenotype. Our results indicate that: (i) the tumor-associated R237C variant is a possible susceptibility factor, but not likely a driver of cancer cell phenotypes, (ii) overexpression of APE1 does not readily promote cellular transformation, and (iii) haploinsufficiency at the APE1 locus can have profound cellular consequences, consistent with BER playing a critical role in proliferating cells. Environ. Mol. Mutagen. 58:84-98, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jennifer L. Illuzzi
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Daniel R. McNeill
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Paul Bastian
- Laboratory of Genetics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Boris Brenerman
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Robert Wersto
- Flow Cytometry Unit, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Helen R. Russell
- Genetics Department, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Peter J. McKinnon
- Genetics Department, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Kevin G. Becker
- Laboratory of Genetics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - David M. Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| |
Collapse
|
26
|
de Melo JTA, de Souza Timoteo AR, Lajus TBP, Brandão JA, de Souza-Pinto NC, Menck CFM, Campalans A, Radicella JP, Vessoni AT, Muotri AR, Agnez-Lima LF. XPC deficiency is related to APE1 and OGG1 expression and function. Mutat Res 2016; 784-785:25-33. [PMID: 26811994 DOI: 10.1016/j.mrfmmm.2016.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/04/2016] [Accepted: 01/14/2016] [Indexed: 12/11/2022]
Abstract
Oxidative DNA damage is considered to be a major cause of neurodegeneration and internal tumors observed in syndromes that result from nucleotide excision repair (NER) deficiencies, such as Xeroderma Pigmentosum (XP) and Cockayne Syndrome (CS). Recent evidence has shown that NER aids in removing oxidized DNA damage and may interact with base excision repair (BER) enzymes. Here, we investigated APE1 and OGG1 expression, localization and activity after oxidative stress in XPC-deficient cells. The endogenous APE1 and OGG1 mRNA levels were lower in XPC-deficient fibroblasts. However, XPC-deficient cells did not show hypersensitivity to oxidative stress compared with NER-proficient cells. To confirm the impact of an XPC deficiency in regulating APE1 and OGG1 expression and activity, we established an XPC-complemented cell line. Although the XPC complementation was only partial and transient, the transfected cells exhibited greater OGG1 expression and activity compared with XPC-deficient cells. However, the APE1 expression and activity did not significantly change. Furthermore, we observed a physical interaction between the XPC and APE1 proteins. Together, the results indicate that the responses of XPC-deficient cells under oxidative stress may not only be associated with NER deficiency per se but may also include new XPC functions in regulating BER proteins.
Collapse
Affiliation(s)
- Julliane Tamara Araújo de Melo
- Laboratório de Biologia Molecular e Genômica, Departamento de Biologia Celular e Genética, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Ana Rafaela de Souza Timoteo
- Laboratório de Biologia Molecular e Genômica, Departamento de Biologia Celular e Genética, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Tirzah Braz Petta Lajus
- Laboratório de Biologia Molecular e Genômica, Departamento de Biologia Celular e Genética, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Juliana Alves Brandão
- Laboratório de Biologia Molecular e Genômica, Departamento de Biologia Celular e Genética, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Nadja Cristhina de Souza-Pinto
- Laboratório de Genética Mitocondrial, Departamento de Química, Instituto de Química, Universidade de São Paulo-USP, São Paulo, Brazil
| | - Carlos Frederico Martins Menck
- Laboratório de Reparo de DNA, Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo-USP, São Paulo, Brazil
| | - Anna Campalans
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire, 18 Route du Panorama, F-92265 Fontenay aux Roses, France
| | - J Pablo Radicella
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire, 18 Route du Panorama, F-92265 Fontenay aux Roses, France
| | - Alexandre Teixeira Vessoni
- Laboratório de Reparo de DNA, Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo-USP, São Paulo, Brazil; Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Alysson Renato Muotri
- Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Lucymara Fassarella Agnez-Lima
- Laboratório de Biologia Molecular e Genômica, Departamento de Biologia Celular e Genética, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
27
|
Di Maso V, Mediavilla MG, Vascotto C, Lupo F, Baccarani U, Avellini C, Tell G, Tiribelli C, Crocè LS. Transcriptional Up-Regulation of APE1/Ref-1 in Hepatic Tumor: Role in Hepatocytes Resistance to Oxidative Stress and Apoptosis. PLoS One 2015; 10:e0143289. [PMID: 26624999 PMCID: PMC4666459 DOI: 10.1371/journal.pone.0143289] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 11/03/2015] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Human Hepatocellular Carcinoma (HCC) is the fifth most frequent neoplasm worldwide and the most serious complication of long-standing chronic liver diseases (CLD). Its development is associated with chronic inflammation and sustained oxidative stress. Deregulation of apurinic apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1), a master regulator of cellular response to oxidative stress, has been associated with poor prognosis in several cancers including HCC. DESIGN In the present study we investigated the APE1/Ref-1 mRNA levels in cirrhotic and HCC tissues obtained during HCC resection. The possible protective role of APE1/Ref-1 against oxidative stress and apoptosis was evaluated in vitro in immortalized human hepatocytes (IHH) over-expressing APE1/Ref-1. RESULTS APE1/Ref-1 was up-regulated in HCC, regulation occurring at the transcriptional level. APE1/Ref-1 mRNA content increased with the progression of liver disease with the transcriptional up-regulation present in cirrhosis significantly increased in HCC. The up-regulation was higher in the less differentiated cancers. In vitro, over-expression of APE1/Ref-1 in normal hepatocytes conferred cell protection against oxidative stress and it was associated with BAX inhibition and escape from apoptosis. CONCLUSION APE1/Ref-1 is up-regulated in HCC and this over-expression correlates with cancer aggressiveness. The up-regulation occurs at the transcriptional level and it is present in the earliest phases of hepatocarcinogenesis. The APE-1/Ref-1 over-expression is associated with hepatocyte survival and inhibits BAX activation and apoptosis. These data suggest a possible role of APE1/Ref-1 over-expression both in hepatocyte survival and HCC development calling attention to this molecule as a promising marker for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Vittorio Di Maso
- FIF- Fondazione Italiana Fegato Area Science Park Basovizza. Trieste, Italia
| | | | - Carlo Vascotto
- Dipartimento Scienze Mediche e Biologiche, Università di Udine. Udine, Itala
| | - Francesco Lupo
- Chirurgia Generale 2 Centro Trapianto Fegato Universitá Torino. Torino, Italia
| | - Umberto Baccarani
- Dipartimento Scienze Mediche e Biologiche, Università di Udine. Udine, Itala
| | - Claudio Avellini
- Dipartimento Scienze Mediche e Biologiche, Università di Udine. Udine, Itala
| | - Gianluca Tell
- FIF- Fondazione Italiana Fegato Area Science Park Basovizza. Trieste, Italia
- Dipartimento Scienze Mediche e Biologiche, Università di Udine. Udine, Itala
| | - Claudio Tiribelli
- FIF- Fondazione Italiana Fegato Area Science Park Basovizza. Trieste, Italia
- Dipartimento Scienze Mediche, Università di Trieste. Trieste, Italia
| | - Lory Saveria Crocè
- FIF- Fondazione Italiana Fegato Area Science Park Basovizza. Trieste, Italia
- Dipartimento Scienze Mediche, Università di Trieste. Trieste, Italia
| |
Collapse
|
28
|
Sholomskas LM, Roche KL, Bloomer SA. Aging impairs induction of redox factor-1 after heat stress: a potential mechanism for heat-induced liver injury. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2015; 7:14-26. [PMID: 26069525 PMCID: PMC4446385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/18/2015] [Indexed: 06/04/2023]
Abstract
Aging is associated with reduced tolerance to physiological stressors such as hyperthermia. In animal models, heat stress is associated with increased oxidative damage in the livers of old rats. In this study, we evaluated the expression of redox factor-1 (Ref-1), a DNA repair enzyme, and thioredoxin-1 (Trx-1), an antioxidant protein. We hypothesized that these proteins would be induced by heat stress in young animals, and that aging would attenuate this response. Young (6 mo) and old (24 mo) male Fischer 344 rats were exposed to a two-heat stress protocol, and livers were harvested at several time points after the second heat stress. Ref-1 and Trx-1 were evaluated by immunoblot and immunohistochemistry. In young rats, Ref-1 was induced by ~50% immediately (0 h) after heat stress, and returned to control levels at 2 h. We observed no change in Ref-1 after hyperthermia in old rats; however, aging was associated with a 2-fold increase in Ref-1 expression. At 2 h after heat stress, Trx-1 was increased in old rats, but there was no change in young rats. In tissue sections, we observed frequent ductular reactions in the old rats that were positive for both Ref-1 and Trx-1. The impairment in the induction of Ref-1 suggests a mechanism for the increased oxidative injury observed in old rats after heat stress. Furthermore, the observation of ductular reactions positive for both Ref-1 and Trx-1 demonstrates a proliferative cellular niche that develops with aging.
Collapse
Affiliation(s)
- Leslee M Sholomskas
- Division of Science and Engineering, Penn State Abington College Abington PA, 19001
| | - Kathryn L Roche
- Division of Science and Engineering, Penn State Abington College Abington PA, 19001
| | - Steven A Bloomer
- Division of Science and Engineering, Penn State Abington College Abington PA, 19001
| |
Collapse
|
29
|
Long-term arsenite exposure induces premature senescence in B cell lymphoma A20 cells. Arch Toxicol 2015; 90:793-803. [PMID: 25787150 DOI: 10.1007/s00204-015-1500-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 03/05/2015] [Indexed: 12/29/2022]
Abstract
Chronic arsenite exposure induces immunosuppression, but the precise mechanisms remain elusive. Our previous studies demonstrated that arsenite exposure for 24 h induces G0/G1 arrest in mouse B lymphoma A20 cells and the arrest is caused through induction of cyclin-dependent kinase inhibitor p16(INK4a) followed by accumulation of an Rb family protein, p130. In this study, we further investigated the consequences of long-term arsenite exposure of A20 cells. The results demonstrated that exposure to 10 μM sodium arsenite up to 14 days induces a great increase in G0/G1 arrest, irreversible cell growth suppression, cellular morphological changes and positive staining for senescence-associated β-galactosidase. The long-term arsenite exposure also induced up-regulation of p16(INK4a) followed by robust accumulation of p130 and activation of the p53 pathway. Knockdown experiments with siRNA showed that p130 accumulation is essential for cell cycle arrest by long-term arsenite exposure. Since p16(INK4a) and the p53 pathway are known to be activated by DNA damage, we investigated the involvement of DNA damage formation by long-term arsenite exposure. We found that a variety of DNA repair-related genes were significantly down-regulated from 24 h of arsenite exposure and activation-induced cytidine deaminase was greatly up-regulated after long-term arsenite exposure. Consistent with these findings, long-term arsenite exposure increased a DNA double-strand break marker, γ-H2AX and increased mutation frequency in a Bcl6 gene region. These results revealed that long-term arsenite exposure induces premature senescence through DNA damage increase and p130 accumulation in lymphoid cells.
Collapse
|
30
|
Bravard A, Auvré F, Fantini D, Bernardino-Sgherri J, Sissoëff L, Daynac M, Xu Z, Etienne O, Dehen C, Comoy E, Boussin FD, Tell G, Deslys JP, Radicella JP. The prion protein is critical for DNA repair and cell survival after genotoxic stress. Nucleic Acids Res 2014; 43:904-16. [PMID: 25539913 PMCID: PMC4333392 DOI: 10.1093/nar/gku1342] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The prion protein (PrP) is highly conserved and ubiquitously expressed, suggesting that it plays an important physiological function. However, despite decades of investigation, this role remains elusive. Here, by using animal and cellular models, we unveil a key role of PrP in the DNA damage response. Exposure of neurons to a genotoxic stress activates PRNP transcription leading to an increased amount of PrP in the nucleus where it interacts with APE1, the major mammalian endonuclease essential for base excision repair, and stimulates its activity. Preventing the induction of PRNP results in accumulation of abasic sites in DNA and impairs cell survival after genotoxic treatment. Brains from Prnp−/− mice display a reduced APE1 activity and a defect in the repair of induced DNA damage in vivo. Thus, PrP is required to maintain genomic stability in response to genotoxic stresses.
Collapse
Affiliation(s)
- Anne Bravard
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Frédéric Auvré
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Damiano Fantini
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Jacqueline Bernardino-Sgherri
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Ludmilla Sissoëff
- CEA, Institut des Maladies Emergentes et des Thérapies Innovantes, Service d'Etudes des Prions et des Infections Atypiques, F-92265 Fontenay-aux-roses, France
| | - Mathieu Daynac
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Zhou Xu
- CEA, Institut des Maladies Emergentes et des Thérapies Innovantes, Service d'Etudes des Prions et des Infections Atypiques, F-92265 Fontenay-aux-roses, France
| | - Olivier Etienne
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Capucine Dehen
- CEA, Institut des Maladies Emergentes et des Thérapies Innovantes, Service d'Etudes des Prions et des Infections Atypiques, F-92265 Fontenay-aux-roses, France
| | - Emmanuel Comoy
- CEA, Institut des Maladies Emergentes et des Thérapies Innovantes, Service d'Etudes des Prions et des Infections Atypiques, F-92265 Fontenay-aux-roses, France
| | - François D Boussin
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| | - Gianluca Tell
- Department of Medical and Biological Sciences, University of Udine, I-33100 Udine, Italy
| | - Jean-Philippe Deslys
- CEA, Institut des Maladies Emergentes et des Thérapies Innovantes, Service d'Etudes des Prions et des Infections Atypiques, F-92265 Fontenay-aux-roses, France
| | - J Pablo Radicella
- CEA, Institute of Cellular and Molecular Radiobiology, F-92265 Fontenay-aux-Roses, France INSERM, U967, F-92265 Fontenay-aux-Roses, France Université Paris Diderot, UMR 967, F-92265 Fontenay-aux-Roses, France Université Paris Sud, UMR 967, F-92265 Fontenay-aux-Roses, France
| |
Collapse
|
31
|
Brenerman BM, Illuzzi JL, Wilson DM. Base excision repair capacity in informing healthspan. Carcinogenesis 2014; 35:2643-52. [PMID: 25355293 PMCID: PMC4247524 DOI: 10.1093/carcin/bgu225] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 12/21/2022] Open
Abstract
Base excision repair (BER) is a frontline defense mechanism for dealing with many common forms of endogenous DNA damage, several of which can drive mutagenic or cell death outcomes. The pathway engages proteins such as glycosylases, abasic endonucleases, polymerases and ligases to remove substrate modifications from DNA and restore the genome back to its original state. Inherited mutations in genes related to BER can give rise to disorders involving cancer, immunodeficiency and neurodegeneration. Studies employing genetically defined heterozygous (haploinsufficient) mouse models indicate that partial reduction in BER capacity can increase vulnerability to both spontaneous and exposure-dependent pathologies. In humans, measurement of BER variation has been imperfect to this point, yet tools to assess BER in epidemiological surveys are steadily evolving. We provide herein an overview of the BER pathway and discuss the current efforts toward defining the relationship of BER defects with disease susceptibility.
Collapse
Affiliation(s)
- Boris M Brenerman
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jennifer L Illuzzi
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - David M Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
32
|
Zhang JH, Di Y, Wu LY, He YL, Zhao T, Huang X, Ding XF, Wu KW, Fan M, Zhu LL. 5-HMF prevents against oxidative injury via APE/Ref-1. Free Radic Res 2014; 49:86-94. [DOI: 10.3109/10715762.2014.981260] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
New Proteomic Insights on the Role of NPR-A in Regulating Self-Renewal of Embryonic Stem Cells. Stem Cell Rev Rep 2014; 10:561-72. [DOI: 10.1007/s12015-014-9517-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Investigation of the effects of short-term inhalation of carbon nanoparticles on brains and lungs of c57bl/6j and p47(phox-/-) mice. Neurotoxicology 2014; 43:65-72. [PMID: 24792328 DOI: 10.1016/j.neuro.2014.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/28/2014] [Accepted: 04/22/2014] [Indexed: 12/15/2022]
Abstract
Recent studies indicate that the brain is a target for toxic carbonaceous nanoparticles present in ambient air. It has been proposed that the neurotoxic effects of such particles are driven by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase mediated generation of reactive oxygen species (ROS) in activated microglia. In the present study, we have evaluated the effects of short term (4h) nose-only inhalation exposure to carbon NP (CNP) in the brains and lungs of C57BL/6J mice and in p47(phox-/-) mice that lack a functional NADPH oxidase. It was shown that the lungs of the p47(phox-/-) mice are less responsive to CNP inhalation than lungs of the corresponding C57BL/6J control animals. Lung tissue mRNA expression of the oxidative stress/DNA damage response genes 8-oxoguanine glycosylase (OGG1) and apurinic/apyrimidinic endonuclease 1 (APE1) were induced by CNP exposure in C57BL/6J but not in the p47(phox-/-) mice. In contrast, the expression of these genes, as well as Tumor Necrosis Factor-α (TNFα), Cyclooxygenase-2 (COX-2) and Heme Oxygenase-1 (HO-1) was not altered in the olfactory bulb, cerebellum or remaining brain tissue part of either mouse background. This indicates that neuroinflammation was not induced by this exposure. CNP inhalation for 4h or for 4h on three consecutive days also did not affect brain tissue protein expression of interleukin (IL)-1β, while a clear significant difference in constitutive expression level of this pro-inflammatory cytokine was found between C57BL/6J and p47(phox-/-) mice. In conclusion, short-term inhalation exposure to pure carbon nanoparticles can trigger mild p47(phox) dependent oxidative stress responses in the lungs of mice whereas in their brains at the same exposure levels signs of oxidative stress and inflammation remain absent. The possible role of p47(phox) in the neuro-inflammatory effects of nanoparticles in vivo remains to be clarified.
Collapse
|
35
|
Wyatt MD. Advances in understanding the coupling of DNA base modifying enzymes to processes involving base excision repair. Adv Cancer Res 2014; 119:63-106. [PMID: 23870509 DOI: 10.1016/b978-0-12-407190-2.00002-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This chapter describes some of the recent, exciting developments that have characterized and connected processes that modify DNA bases with DNA repair pathways. It begins with AID/APOBEC or TET family members that covalently modify bases within DNA. The modified bases, such as uracil or 5-formylcytosine, are then excised by DNA glycosylases including UNG or TDG to initiate base excision repair (BER). BER is known to preserve genome integrity by removing damaged bases. The newer studies underscore the necessity of BER following enzymes that deliberately damage DNA. This includes the role of BER in antibody diversification and more recently, its requirement for demethylation of 5-methylcytosine in mammalian cells. The recent advances have shed light on mechanisms of DNA demethylation, and have raised many more questions. The potential hazards of these processes have also been revealed. Dysregulation of the activity of base modifying enzymes, and resolution by unfaithful or corrupt means can be a driver of genome instability and tumorigenesis. The understanding of both DNA and histone methylation and demethylation is now revealing the true extent to which epigenetics influence normal development and cancer, an abnormal development.
Collapse
Affiliation(s)
- Michael D Wyatt
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA.
| |
Collapse
|
36
|
Abstract
SIGNIFICANCE Human apurinic/apyrimidinic endonuclease 1 (APE1, also known as REF-1) was isolated based on its ability to cleave at AP sites in DNA or activate the DNA binding activity of certain transcription factors. We review herein topics related to this multi-functional DNA repair and stress-response protein. RECENT ADVANCES APE1 displays homology to Escherichia coli exonuclease III and is a member of the divalent metal-dependent α/β fold-containing phosphoesterase superfamily of enzymes. APE1 has acquired distinct active site and loop elements that dictate substrate selectivity, and a unique N-terminus which at minimum imparts nuclear targeting and interaction specificity. Additional activities ascribed to APE1 include 3'-5' exonuclease, 3'-repair diesterase, nucleotide incision repair, damaged or site-specific RNA cleavage, and multiple transcription regulatory roles. CRITICAL ISSUES APE1 is essential for mouse embryogenesis and contributes to cell viability in a genetic background-dependent manner. Haploinsufficient APE1(+/-) mice exhibit reduced survival, increased cancer formation, and cellular/tissue hyper-sensitivity to oxidative stress, supporting the notion that impaired APE1 function associates with disease susceptibility. Although abnormal APE1 expression/localization has been seen in cancer and neuropathologies, and impaired-function variants have been described, a causal link between an APE1 defect and human disease remains elusive. FUTURE DIRECTIONS Ongoing efforts aim at delineating the biological role(s) of the different APE1 activities, as well as the regulatory mechanisms for its intra-cellular distribution and participation in diverse molecular pathways. The determination of whether APE1 defects contribute to human disease, particularly pathologies that involve oxidative stress, and whether APE1 small-molecule regulators have clinical utility, is central to future investigations.
Collapse
Affiliation(s)
- Mengxia Li
- Intramural Research Program, Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | | |
Collapse
|
37
|
Groenendyk J, Agellon LB, Michalak M. Coping with endoplasmic reticulum stress in the cardiovascular system. Annu Rev Physiol 2012; 75:49-67. [PMID: 23020580 DOI: 10.1146/annurev-physiol-030212-183707] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The endoplasmic reticulum (ER) is a multifunctional intracellular organelle, a component of the cellular reticular network that allows cells to adjust to a wide variety of conditions. The cardiomyocyte reticular network is the ideal location of sensors for both intrinsic and extrinsic factors that disrupt energy and/or nutrient homeostasis and lead to ER stress, a disturbance in ER function. ER stress has been linked to both physiological and pathological states in the cardiovascular system; such states include myocardial infarction, oxygen starvation (hypoxia) and fuel starvation, ischemia, pressure overload, dilated cardiomyopathy, hypertrophy, and heart failure. The ER stress coping response (e.g., the unfolded protein response) is composed of discrete pathways that are controlled by a collection of common regulatory components that may function as a single entity involved in reacting to ER stress. These corrective strategies allow the cardiomyocyte reticular network to restore energy and/or nutrient homeostasis and to avoid cell death. Therefore, the identities of the ER stress corrective strategies are important targets for the development of therapeutic approaches for cardiovascular and other acquired disorders.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
38
|
Tasaki M, Kuroiwa Y, Inoue T, Hibi D, Matsushita K, Ishii Y, Maruyama S, Nohmi T, Nishikawa A, Umemura T. Oxidative DNA damage andin vivomutagenicity caused by reactive oxygen species generated in the livers ofp53-proficient or -deficientgptdelta mice treated with non-genotoxic hepatocarcinogens. J Appl Toxicol 2012; 33:1433-41. [DOI: 10.1002/jat.2807] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/28/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Affiliation(s)
- Masako Tasaki
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Yuichi Kuroiwa
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Tomoki Inoue
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Daisuke Hibi
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Kohei Matsushita
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Yuji Ishii
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Soichi Maruyama
- Laboratory of Veterinary Public Health, Department of Veterinary Medicine, College of Bioresource Science; Nihon University; 1866, Kameino Fujisawa-city Kanagawa 252-8510 Japan
| | - Takehiko Nohmi
- Division of Genetics and Mutagenesis; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Akiyoshi Nishikawa
- Biological Safety Research Center; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| | - Takashi Umemura
- Division of Pathology; National Institute of Health Sciences; 1-18-1, Kamiyoga, Setagaya-ku Tokyo 158-8501 Japan
| |
Collapse
|
39
|
DNA Repair and Cancer Therapy: Targeting APE1/Ref-1 Using Dietary Agents. JOURNAL OF ONCOLOGY 2012; 2012:370481. [PMID: 22997517 PMCID: PMC3444914 DOI: 10.1155/2012/370481] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 07/31/2012] [Indexed: 12/21/2022]
Abstract
Epidemiological studies have demonstrated the cancer protective effects of dietary agents and other natural compounds isolated from fruits, soybeans, and vegetables on neoplasia. Studies have also revealed the potential for these natural products to be combined with chemotherapy or radiotherapy for the more effective treatment of cancer. In this paper we discuss the potential for targeting the DNA base excision repair enzyme APE1/Ref-1 using dietary agents such as soy isoflavones, resveratrol, curcumin, and the vitamins ascorbate and α-tocopherol. We also discuss the potential role of soy isoflavones in sensitizing cancer cells to the effects of radiotherapy. A comprehensive review of the dual nature of APE1/Ref-1 in DNA repair and redox activation of cellular transcription factors, NF-κB and HIF-1α, is also discussed. Further research efforts dedicated to delineating the role of APE1/Ref-1 DNA repair versus redox activity in sensitizing cancer cells to conventional treatment are warranted.
Collapse
|
40
|
Leite de Oliveira R, Deschoemaeker S, Henze AT, Debackere K, Finisguerra V, Takeda Y, Roncal C, Dettori D, Tack E, Jönsson Y, Veschini L, Peeters A, Anisimov A, Hofmann M, Alitalo K, Baes M, D'hooge J, Carmeliet P, Mazzone M. Gene-targeting of Phd2 improves tumor response to chemotherapy and prevents side-toxicity. Cancer Cell 2012; 22:263-77. [PMID: 22897855 DOI: 10.1016/j.ccr.2012.06.028] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 05/08/2012] [Accepted: 06/26/2012] [Indexed: 12/18/2022]
Abstract
The success of chemotherapy in cancer treatment is limited by scarce drug delivery to the tumor and severe side-toxicity. Prolyl hydroxylase domain protein 2 (PHD2) is an oxygen/redox-sensitive enzyme that induces cellular adaptations to stress conditions. Reduced activity of PHD2 in endothelial cells normalizes tumor vessels and enhances perfusion. Here, we show that tumor vessel normalization by genetic inactivation of Phd2 increases the delivery of chemotherapeutics to the tumor and, hence, their antitumor and antimetastatic effect, regardless of combined inhibition of Phd2 in cancer cells. In response to chemotherapy-induced oxidative stress, pharmacological inhibition or genetic inactivation of Phd2 enhances a hypoxia-inducible transcription factor (HIF)-mediated detoxification program in healthy organs, which prevents oxidative damage, organ failure, and tissue demise. Altogether, our study discloses alternative strategies for chemotherapy optimization.
Collapse
|
41
|
Li M, Vascotto C, Xu S, Dai N, Qing Y, Zhong Z, Tell G, Wang D. Human AP endonuclease/redox factor APE1/ref-1 modulates mitochondrial function after oxidative stress by regulating the transcriptional activity of NRF1. Free Radic Biol Med 2012; 53:237-48. [PMID: 22580151 DOI: 10.1016/j.freeradbiomed.2012.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/06/2012] [Accepted: 04/06/2012] [Indexed: 12/11/2022]
Abstract
Maintenance of mitochondrial functionality largely depends on nuclear transcription because most mitochondrial proteins are encoded by the nuclear genome and transported to the mitochondria. Nuclear respiration factor 1 (NRF1) plays a crucial role in regulating the expression of a broad range of mitochondrial genes in the nucleus in response to cellular oxidative stress. However, little is known about the redox regulatory mechanism of the transcriptional activity of NRF1. In this study, we show that the human apurinic/apyrimidinic endonuclease/redox factor (APE1/Ref-1) is involved in mitochondrial function regulation by modulating the DNA-binding activity of NRF1. Our results show that both APE1 expression level and its redox activity are essential for maintenance of the mitochondrial function after tert-butylhydroperoxide-induced oxidative stress. Upon knocking down or redox mutation of APE1, NRF1 DNA-binding activity was impaired and, consequently, the expression of its downstream genes, including Tfam, Cox6c, and Tomm22, was significantly reduced. NRF1 knockdown blocked the restoration of mitochondrial function by APE1 overexpression, which further suggests APE1 regulates mitochondrial function through an NRF1-dependent pathway. Taken together, our results reveal APE1 as a new coactivator of NRF1, which highlights an additional regulatory role of APE1 in maintenance of mitochondrial functionality.
Collapse
Affiliation(s)
- Mengxia Li
- Cancer Center and Department of Pathology, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhang L, Yesupriya A, Hu DJ, Chang MH, Dowling NF, Ned RM, Udhayakumar V, Lindegren ML, Khudyakov Y. Variants in ABCB1, TGFB1, and XRCC1 genes and susceptibility to viral hepatitis A infection in Mexican Americans. Hepatology 2012; 55:1008-18. [PMID: 22135187 DOI: 10.1002/hep.25513] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
UNLABELLED Hepatitis A vaccination has dramatically reduced the incidence of hepatitis A virus (HAV) infection, but new infections continue to occur. To identify human genetic variants conferring a risk for HAV infection among the three major racial/ethnic populations in the United States, we assessed associations between 67 genetic variants (single nucleotide polymorphisms [SNPs]) among 31 candidate genes and serologic evidence of prior HAV infection using a population-based, cross-sectional study of 6,779 participants, including 2,619 non-Hispanic whites, 2,095 non-Hispanic blacks, and 2,065 Mexican Americans enrolled in phase 2 (1991-1994) of the Third National Health and Nutrition Examination Survey. Among the three racial/ethnic groups, the number (weighted frequency) of seropositivity for antibody to HAV was 958 (24.9%), 802 (39.2%), and 1540 (71.5%), respectively. No significant associations with any of the 67 SNPs were observed among non-Hispanic whites or non-Hispanic blacks. In contrast, among Mexican Americans, variants in two genes were found to be associated with an increased risk of HAV infection: TGFB1 rs1800469 (adjusted odds ratio [OR], 1.38; 95% confidence interval [CI], 1.14-1.68; P value adjusted for false discovery rate [FDR-P] = 0.017) and XRCC1 rs1799782 (OR, 1.57; 95% CI, 1.27-1.94; FDR-P = 0.0007). A decreased risk was found with ABCB1 rs1045642 (OR, 0.79; 95% CI, 0.71-0.89; FDR-P = 0.0007). CONCLUSION Genetic variants in ABCB1, TGFB1, and XRCC1 appear to be associated with susceptibility to HAV infection among Mexican Americans. Replication studies involving larger population samples are warranted.
Collapse
Affiliation(s)
- Lyna Zhang
- Office of Public Health Genomics, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Diseases Control and Prevention, 1600 Clifton Road NE, Atlanta, GA 30333, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Robbins D, Li W, Humphrey K, Zhao Y. Redox Factor-1 Mediates Inflammatory Response during Tumor Promotion in Skin Epidermal JB6 Cells. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/ojapo.2012.13003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
44
|
Meisenberg C, Tait PS, Dianova II, Wright K, Edelmann MJ, Ternette N, Tasaki T, Kessler BM, Parsons JL, Kwon YT, Dianov GL. Ubiquitin ligase UBR3 regulates cellular levels of the essential DNA repair protein APE1 and is required for genome stability. Nucleic Acids Res 2011; 40:701-11. [PMID: 21933813 PMCID: PMC3258136 DOI: 10.1093/nar/gkr744] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
APE1 (Ref-1) is an essential human protein involved in DNA damage repair and regulation of transcription. Although the cellular functions and biochemical properties of APE1 are well characterized, the mechanism involved in regulation of the cellular levels of this important DNA repair/transcriptional regulation enzyme, remains poorly understood. Using an in vitro ubiquitylation assay, we have now purified the human E3 ubiquitin ligase UBR3 as a major activity that polyubiquitylates APE1 at multiple lysine residues clustered on the N-terminal tail. We further show that a knockout of the Ubr3 gene in mouse embryonic fibroblasts leads to an up-regulation of the cellular levels of APE1 protein and subsequent genomic instability. These data propose an important role for UBR3 in the control of the steady state levels of APE1 and consequently error free DNA repair.
Collapse
Affiliation(s)
- Cornelia Meisenberg
- Gray Institute for Radiation Oncology and Biology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Unnikrishnan A, Prychitko TM, Patel HV, Chowdhury ME, Pilling AB, Ventrella-Lucente LF, Papakonstantinou EV, Cabelof DC, Heydari AR. Folate deficiency regulates expression of DNA polymerase β in response to oxidative stress. Free Radic Biol Med 2011; 50:270-80. [PMID: 21070850 PMCID: PMC3018545 DOI: 10.1016/j.freeradbiomed.2010.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 10/08/2010] [Accepted: 11/01/2010] [Indexed: 01/19/2023]
Abstract
Folate deficiency has been shown to influence carcinogenesis by creating an imbalance in the base excision repair (BER) pathway, affecting BER homeostasis. The inability to mount a BER response to oxidative stress in a folate-deficient environment results in the accumulation of DNA repair intermediates, i.e., DNA strand breaks. Our data indicate that upregulation of β-pol expression in response to oxidative stress is inhibited by folate deficiency at the level of gene expression. Alteration in the expression of β-pol in a folate-deficient environment is not due to epigenetic changes in the core promoter of the β-pol gene, i.e., the CpG islands within the β-pol promoter remain unmethylated in the presence or absence of folate. However, the promoter analysis studies show a differential binding of regulatory factors to the -36 to -7 region (the folic acid-response region, FARR) within the core promoter of β-pol. Moreover, we observe a tight correlation between the level of binding of regulatory factors with the FARR and inhibition of β-pol expression. Based on these findings, we propose that folate deficiency results in an upregulation/stability of negative regulatory factors interacting with FARR, repressing the upregulation of the β-pol gene in response to oxidative stress.
Collapse
Affiliation(s)
- Archana Unnikrishnan
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Tom M. Prychitko
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Hiral V. Patel
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Mahbuba E. Chowdhury
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Amanda B. Pilling
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Lisa F. Ventrella-Lucente
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Erin V. Papakonstantinou
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
| | - Diane C. Cabelof
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
- Barbara Ann Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan 48202
| | - Ahmad R. Heydari
- Department of Nutrition and Food science, Science College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan 48202
- Barbara Ann Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan 48202
| |
Collapse
|
46
|
van Berlo D, Wessels A, Boots AW, Wilhelmi V, Scherbart AM, Gerloff K, van Schooten FJ, Albrecht C, Schins RPF. Neutrophil-derived ROS contribute to oxidative DNA damage induction by quartz particles. Free Radic Biol Med 2010; 49:1685-93. [PMID: 20828610 DOI: 10.1016/j.freeradbiomed.2010.08.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 08/09/2010] [Accepted: 08/29/2010] [Indexed: 11/26/2022]
Abstract
The carcinogenicity of respirable quartz is considered to be driven by reactive oxygen species (ROS) generation in association with chronic inflammation. The contribution of phagocyte-derived ROS to inflammation, oxidative stress, and DNA damage responses was investigated in the lungs of C57BL/6J wild-type and p47(phox-/-) mice, 24h after pharyngeal aspiration of DQ12 quartz (100 mg/kg bw). Bone-marrow-derived neutrophils from wild-type and p47(phox-/-) mice were used for parallel in vitro investigations in coculture with A549 human alveolar epithelial cells. Quartz induced a marked neutrophil influx in both wild-type and p47(phox-/-) mouse lungs. Significant increases in mRNA expression of the oxidative stress markers HO-1 and γ-GCS were observed only in quartz-treated wild-type animals. Oxidative DNA damage in lung tissue was not affected by quartz exposure and did not differ between p47(phox-/-) and WT mice. Differences in mRNA expression of the DNA repair genes OGG1, APE-1, DNA Polβ, and XRCC1 were also absent. Quartz treatment of cocultures containing wild-type neutrophils, but not p47(phox-/-) neutrophils, caused increased oxidative DNA damage in epithelial cells. Our study demonstrates that neutrophil-derived ROS significantly contribute to pulmonary oxidative stress responses after acute quartz exposure, yet their role in the associated induction of oxidative DNA damage could be shown only in vitro.
Collapse
Affiliation(s)
- Damien van Berlo
- Institut für Umweltmedizinische Forschung, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wessels A, Van Berlo D, Boots AW, Gerloff K, Scherbart AM, Cassee FR, Gerlofs-Nijland ME, Van Schooten FJ, Albrecht C, Schins RPF. Oxidative stress and DNA damage responses in rat and mouse lung to inhaled carbon nanoparticles. Nanotoxicology 2010; 5:66-78. [PMID: 21417689 DOI: 10.3109/17435390.2010.494773] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We have investigated whether short-term nose-only inhalation exposure to electric spark discharge-generated carbon nanoparticles (∼60 nm) causes oxidative stress and DNA damage responses in the lungs of rats (152 μg/m(3); 4 h) and mice (142 μg/m(3); 4 h, or three times 4 h). In both species, no pulmonary inflammation and toxicity were detected by bronchoalveolar lavage or mRNA expression analyses. Oxidative DNA damage (measured by fpg-comet assay), was also not increased in mouse whole lung tissue or isolated lung epithelial cells from rat. In addition, the mRNA expressions of the DNA base excision repair genes OGG1, DNA Polβ and XRCC1 were not altered. However, in the lung epithelial cells isolated from the nanoparticle-exposed rats a small but significant increase in APE-1 mRNA expression was measured. Thus, short-term inhalation of carbon nanoparticles under the applied exposure regimen, does not cause oxidative stress and DNA damage in the lungs of healthy mice and rats.
Collapse
Affiliation(s)
- Anton Wessels
- Institut für Umweltmedizinische Forschung (IUF) an der Heinrich Heine Universität Düsseldorf gGmbH, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ventrella-Lucente LF, Unnikrishnan A, Pilling AB, Patel HV, Kushwaha D, Dombkowski AA, Schmelz EM, Cabelof DC, Heydari AR. Folate deficiency provides protection against colon carcinogenesis in DNA polymerase beta haploinsufficient mice. J Biol Chem 2010; 285:19246-58. [PMID: 20404327 DOI: 10.1074/jbc.m109.069807] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aging and DNA polymerase beta deficiency (beta-pol(+/-)) interact to accelerate the development of malignant lymphomas and adenocarcinoma and increase tumor bearing load in mice. Folate deficiency (FD) has been shown to induce DNA damage repaired via the base excision repair (BER) pathway. We anticipated that FD and BER deficiency would interact to accelerate aberrant crypt foci (ACF) formation and tumor development in beta-pol haploinsufficient animals. FD resulted in a significant increase in ACF formation in wild type (WT) animals exposed to 1,2-dimethylhydrazine, a known colon and liver carcinogen; however, FD reduced development of ACF in beta-pol haploinsufficient mice. Prolonged feeding of the FD diet resulted in advanced ACF formation and liver tumors in wild type mice. However, FD attenuated onset and progression of ACF and prevented liver tumorigenesis in beta-pol haploinsufficient mice, i.e. FD provided protection against tumorigenesis in a BER-deficient environment in all tissues where 1,2-dimethylhydrazine exerts its damage. Here we show a distinct down-regulation in DNA repair pathways, e.g. BER, nucleotide excision repair, and mismatch repair, and decline in cell proliferation, as well as an up-regulation in poly(ADP-ribose) polymerase, proapoptotic genes, and apoptosis in colons of FD beta-pol haploinsufficient mice.
Collapse
Affiliation(s)
- Lisa F Ventrella-Lucente
- Department of Nutrition and Food, Science College of Liberal Arts and Sciences, School of Medicine, Wayne State University, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Selective inhibition of hypoxia-inducible factor (HIF) prolyl-hydroxylase 1 mediates neuroprotection against normoxic oxidative death via HIF- and CREB-independent pathways. J Neurosci 2009; 29:8828-38. [PMID: 19587290 DOI: 10.1523/jneurosci.1779-09.2009] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress contributes to tissue injury in conditions ranging from cardiovascular disease to stroke, spinal cord injury, neurodegeneration, and perhaps even aging. Yet the efficacy of antioxidants in human disease has been mixed at best. We need a better understanding of the mechanisms by which established antioxidants combat oxidative stress. Iron chelators are well established inhibitors of oxidative death in both neural and non-neural tissues, but their precise mechanism of action remains elusive. The prevailing but not completely substantiated view is that iron chelators prevent oxidative injury by suppressing Fenton chemistry and the formation of highly reactive hydroxyl radicals. Here, we show that iron chelation protects, rather unexpectedly, by inhibiting the hypoxia-inducible factor prolyl 4-hydroxylase isoform 1 (PHD1), an iron and 2-oxoglutarate-dependent dioxygenase. PHD1 and its isoforms 2 and 3 are best known for stabilizing transcriptional regulators involved in hypoxic adaptation, such as HIF-1alpha and cAMP response element-binding protein (CREB). Yet we find that global hypoxia-inducible factor (HIF)-PHD inhibition protects neurons even when HIF-1alpha and CREB are directly suppressed. Moreover, two global HIF-PHD inhibitors continued to be neuroprotective even in the presence of diminished HIF-2alpha levels, which itself increases neuronal susceptibility to oxidative stress. Finally, RNA interference to PHD1 but not isoforms PHD2 or PHD3 prevents oxidative death, independent of HIF activation. Together, these studies suggest that iron chelators can prevent normoxic oxidative neuronal death through selective inhibition of PHD1 but independent of HIF-1alpha and CREB; and that HIF-2alpha, not HIF-1alpha, regulates susceptibility to normoxic oxidative neuronal death.
Collapse
|