1
|
Wang J, Chang X, Li C, Gao J, Guo Z, Zhuang H, Wang L, Huang Y, Wang W, Li C, He Q. DNA-PKcs-Driven YAP1 Phosphorylation and Nuclear Translocation: a Key Regulator of Ferroptosis in Hyperglycemia-Induced Cardiac Dysfunction in Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412698. [PMID: 40279648 DOI: 10.1002/advs.202412698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/12/2025] [Indexed: 04/27/2025]
Abstract
The DNA-Dependent Protein Kinase catalytic subunit (DNA-PKcs) acts as a principal executor in the DNA damage response (DDR), mediating the phosphorylation of a broad spectrum of substrates integral to DNA repair and apoptosis. This investigation seeks to discern the possible association and mechanisms linking hyperglycemia-induced ferroptosis and DNA-PKcs in DCM. This data exhibits a substantial activation of DNAPKcs- dependent DDR in mice with streptozotocin-induced DCM. However, deletion of DNA-PKcs in cardiomyocytes notably mitigates DNA damage, enhances heart function and dampens the inflammatory response. Co-IP/MS analysis and subsequent validation experiments demonstrate that DNA-PKcs directly interacts with and phosphorylates YAP1 at Thr226. This phosphorylation event facilitates the nuclear retention of YAP1, where it intensifies the transcription of ferroptosis-associated genes. Knockin mice expressing a nonphosphorylatable T226A YAP1 mutant display decreased ferroptosis, reduced myocardial fibrosis and improved heart function. Taken together, this study unravels that DDR acts as an intracellular stress damage sensor, perceiving hyperglycemic conditions and subsequently transmitting the damage signal to incite ferroptosis through the interplay between DNA-PKcs and YAP1. This novel insight suggests that the DNA-PKcs-mediated YAP1 phosphorylation and the ferroptosis activation could be the promising therapeutic targets for the management of DCM.
Collapse
Affiliation(s)
- Junyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Chun Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jing Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Haowen Zhuang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lingjun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yusheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wei Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Chao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Qingyong He
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
2
|
Jain A, Keesari PR, Pulakurthi YS, Katamreddy R, Dhar M, Desai R. Pancreatic Cancer Risk in Prediabetes: A Systematic Meta-analysis Approach. Pancreas 2025; 54:e51-e56. [PMID: 39324961 DOI: 10.1097/mpa.0000000000002391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
OBJECTIVES Pancreatic cancer and prediabetes pose significant public health challenges. Given the lack of strong evidence we performed a meta-analysis to assess the risk of pancreatic cancer in prediabetes. MATERIALS AND METHODS We performed a thorough search of the major databases over the last 10 years to identify relevant articles. The pooled odds ratio (OR) and hazard ratio (HR) were combined to calculate the effect size (ES). RESULTS We analyzed 5 studies including 5,425,111 prediabetic individuals and 16,096,467 normoglycemic population across 5 countries with a median follow-up of 8.5 years. We identified a noteworthy association between prediabetes and pancreatic cancer, reporting an unadjusted ES of 1.36 (95% confidence interval [CI] 1.05-1.77, P = 0.02) and an adjusted ES of 1.40 (1.23-1.59, P < 0.01). Subgroup analyses by age revealed variations in risk, with studies involving participants aged 60 and above exhibiting a higher ES (ES 1.83, 95% CI 1.28-2.62, P < 0.01). Geographical differences were also observed, with Japanese studies reporting a higher risk (ES 1.89, 95% CI 1.15-3.10, P < 0.01) compared with those from the United States (ES 1.32, 95% CI 1.13-1.53, P < 0.01). CONCLUSIONS We identified 40% higher risk of pancreatic cancer in patients with prediabetes than those with normal blood glucose necessitating urgent attention for further research and policy change.
Collapse
Affiliation(s)
- Akhil Jain
- From the University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Meekoo Dhar
- Staten Island University Hospital, Staten Island, NY
| | | |
Collapse
|
3
|
Ahmed I, Chakraborty R, Faizy AF, Moin S. Exploring the key role of DNA methylation as an epigenetic modulator in oxidative stress related islet cell injury in patients with type 2 diabetes mellitus: a review. J Diabetes Metab Disord 2024; 23:1699-1718. [PMID: 39610516 PMCID: PMC11599646 DOI: 10.1007/s40200-024-01496-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/21/2024] [Indexed: 11/30/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial metabolic disorder characterised by impaired insulin secretion and action, often exacerbated by oxidative stress. Recent research has highlighted the intricate involvement of epigenetic mechanisms, particularly DNA methylation, in the pathogenesis of T2DM. This review aims to elucidate the role of DNA methylation as an epigenetic modifier in oxidative stress-mediated beta cell dysfunction, a key component of T2DM pathophysiology. Oxidative stress, arising from an imbalance between reactive oxygen species (ROS) production and antioxidant defence mechanisms, is a hallmark feature of T2DM. Beta cells, responsible for insulin secretion, are particularly vulnerable to oxidative damage due to their low antioxidant capacity. Emerging evidence suggests that oxidative stress can induce aberrant DNA methylation patterns in beta cells, leading to altered gene expression profiles associated with insulin secretion and cell survival. Furthermore, studies have identified specific genes involved in beta cell function and survival that undergo DNA methylation changes in response to oxidative stress in T2DM. These epigenetic modifications can perpetuate beta cell dysfunction by dysregulating key pathways essential for insulin secretion, such as the insulin signalling cascade and mitochondrial function. Understanding the interplay between DNA methylation, oxidative stress, and beta cell dysfunction holds promise for developing novel therapeutic strategies for T2DM. Targeting aberrant DNA methylation patterns may offer new avenues for restoring beta cell function and improving glycemic control in patients with T2DM. However, further research is needed to elucidate the complex mechanisms underlying epigenetic regulation in T2DM and to translate these findings into clinical interventions.
Collapse
Affiliation(s)
- Istiaque Ahmed
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Ritoja Chakraborty
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Abul Faiz Faizy
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Shagufta Moin
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College and Hospital Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| |
Collapse
|
4
|
Keesari PR, Jain A, Ganampet NR, Subhasri GSD, Edusa S, Muslehuddin Z, Theik NWY, Palisetti S, Salibindla D, Manaktala PS, Desai R. Association between prediabetes and breast cancer: a comprehensive meta-analysis. Breast Cancer Res Treat 2024; 204:1-13. [PMID: 38060076 DOI: 10.1007/s10549-023-07181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Breast cancer accounts for up to 30% of cancer cases in women in the US. Diabetes mellitus has been recognized as a risk factor for breast cancer. Some studies have suggested that prediabetes may also be associated with breast cancer whereas other studies have shown no or an inverse association; thus, we conducted a meta-analysis to assess the risk of breast cancer in prediabetes. METHODS We searched PubMed/Medline, EMBASE, Google Scholar, and Scopus to identify studies that reported breast cancer risks in patients having prediabetes compared to normoglycemic patients. Binary random-effects model was used to calculate a pooled odds ratio (OR) with 95% confidence intervals. I2 statistics were used to assess heterogeneity. Leave-one-out sensitivity analysis and subgroup analyses were performed. RESULTS We analyzed 7 studies with 24,586 prediabetic and 224,314 normoglycemic individuals (783 and 5739 breast cancer cases, respectively). Unadjusted odds ratio (OR) for breast cancer was 1.45 (95% CI = 1.14, 1.83); adjusted OR was 1.19 (95% CI = 1.07, 1.34) in prediabetes. Subgroup analysis revealed a higher breast cancer risk in individuals aged less than 60 years (OR = 1.86, 95% CI = 1.39, 2.49) than in those aged 60 years or more (OR = 1.07, 95% CI = 0.97, 1.18). Subgroup analysis by median follow-up length indicated a higher risk of breast cancer for follow-ups of less than or equal to 2 years (OR = 2.34, 95% CI = 1.85, 2.95) than in those of over 10 years (OR = 1.1, 95% CI = 0.99, 1.23) and 6 to 10 years (OR = 1.03, 95% CI = 0.88, 1.21). CONCLUSIONS In conclusion, individuals with prediabetes have higher risk of developing breast cancer than those with normoglycemia, especially younger prediabetes patients. These individuals may benefit from early identification, monitoring, and interventions to reverse prediabetes.
Collapse
Affiliation(s)
- Praneeth Reddy Keesari
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Akhil Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | | | | | - Samuel Edusa
- Department of Internal Medicine, Piedmont Athens Regional, Athens, GA, USA
| | - Zainab Muslehuddin
- Department of Internal Medicine, Detroit Medical Center, Wayne State University-Sinai Grace Hospital, Detroit, MI, USA
| | | | - Spandana Palisetti
- Department of Medicine, Jawaharlal Nehru Medical College, Belgaum, Karnataka, India
| | | | | | - Rupak Desai
- Independent Researcher, Outcomes Research, Atlanta, GA, USA
| |
Collapse
|
5
|
Yousefzadeh MJ, Huerta Guevara AP, Postmus AC, Flores RR, Sano T, Jurdzinski A, Angelini L, McGowan SJ, O’Kelly RD, Wade EA, Gonzalez-Espada LV, Henessy-Wack D, Howard S, Rozgaja TA, Trussoni CE, LaRusso NF, Eggen BJ, Jonker JW, Robbins PD, Niedernhofer LJ, Kruit JK. Failure to repair endogenous DNA damage in β-cells causes adult-onset diabetes in mice. AGING BIOLOGY 2023; 1:20230015. [PMID: 38124711 PMCID: PMC10732477 DOI: 10.59368/agingbio.20230015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Age is the greatest risk factor for the development of type 2 diabetes mellitus (T2DM). Age-related decline in organ function is attributed to the accumulation of stochastic damage, including damage to the nuclear genome. Islets of T2DM patients display increased levels of DNA damage. However, whether this is a cause or consequence of the disease has not been elucidated. Here, we asked if spontaneous, endogenous DNA damage in β-cells can drive β-cell dysfunction and diabetes, via deletion of Ercc1, a key DNA repair gene, in β-cells. Mice harboring Ercc1-deficient β-cells developed adult-onset diabetes as demonstrated by increased random and fasted blood glucose levels, impaired glucose tolerance, and reduced insulin secretion. The inability to repair endogenous DNA damage led to an increase in oxidative DNA damage and apoptosis in β-cells and a significant loss of β-cell mass. Using electron microscopy, we identified β-cells in clear distress that showed an increased cell size, enlarged nuclear size, reduced number of mature insulin granules, and decreased number of mitochondria. Some β-cells were more affected than others consistent with the stochastic nature of spontaneous DNA damage. Ercc1-deficiency in β-cells also resulted in loss of β-cell function as glucose-stimulated insulin secretion and mitochondrial function were impaired in islets isolated from mice harboring Ercc1-deficient β-cells. These data reveal that unrepaired endogenous DNA damage is sufficient to drive β-cell dysfunction and provide a mechanism by which age increases the risk of T2DM.
Collapse
Affiliation(s)
- Matthew J. Yousefzadeh
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Ana P. Huerta Guevara
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrea C. Postmus
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rafael R. Flores
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Tokio Sano
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Angelika Jurdzinski
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luise Angelini
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Sara J. McGowan
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Ryan D. O’Kelly
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Erin A. Wade
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Lisa V. Gonzalez-Espada
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Danielle Henessy-Wack
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Shannon Howard
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Tania A. Rozgaja
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
| | - Christy E. Trussoni
- Division of Gastroenterology and Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA
| | - Bart J.L. Eggen
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W. Jonker
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul D. Robbins
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Laura J. Niedernhofer
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute, 130 Scripps Way #3B3, Jupiter FL, 33458, USA
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA
| | - Janine K. Kruit
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Wu Y, Deng Y, Dai Z, Ma Y, Lyu L, Lei C, Zheng Y, Li Y, Wang Z, Gao J. Estimates of bladder cancer burden attributable to high fasting plasma glucose: Findings of the Global Burden of Disease Study 2019. Cancer Med 2023; 12:16469-16481. [PMID: 37350559 PMCID: PMC10469723 DOI: 10.1002/cam4.6219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND High fasting plasma glucose (FPG) has been listed as one of the risk factors for bladder cancer. We here estimated the global, regional, and national levels of bladder cancer burden attributable to high FPG from 1990 to 2019. METHODS Bladder cancer data attributable to high FPG were extracted from the Global Burden of Disease Study 2019, and analyzed by age, sex, year, and location. Age-standardized rates were utilized to evaluate the burden between different populations. The temporal trend of the burden was estimated through the Joinpoint analysis. RESULTS In 2019, high FPG contributed to 22,823.33 (95% uncertainty interval [UI], 4694.88-48,962.26) deaths and 399,654.91 (95% UI, 81,609.35-865,890.95) disability-adjusted life years (DALYs) of bladder cancer globally. Since 1990, the global age-standardized death and DALY rates of bladder cancer attributable to high FPG increased apparently by 39.18% and 41.48%, respectively. During the last 30 years, high FPG-related age-standardized death and DALY rates of bladder cancer have increased in all countries. In 2019, Central Europe showed the greatest high FPG-related age-standardized death and DALY rates of bladder cancer, but Andean Latin America had the lowest rates. Nationally, Lebanon showed the greatest high FPG-related age-standardized death and DALY rates of bladder cancer in 2019. High FPG-attributable deaths and DALYs of bladder cancer were more considerable among males and older people. Countries with high SDI showed higher levels of age-standardized death and DALY rates of bladder cancer due to high FPG and presented remarkable upward trends in rates in the last 30 years. CONCLUSIONS Globally, the high FPG-associated bladder cancer burden has remarkably increased in all countries, and showed a higher level among countries with higher SDI. Monitoring FPG levels among patients with bladder cancer is critical to lower the corresponding burden.
Collapse
Affiliation(s)
- Ying Wu
- Department of UrologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yujiao Deng
- Department of NephrologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Zhijun Dai
- Department of Breast SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yubo Ma
- Department of UrologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Lijuan Lyu
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Chen Lei
- Department of EndocrinologyThe General Hospital of Ningxia Medical UniversityYinchuanChina
| | - Yi Zheng
- Department of NephrologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yizhen Li
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Ziming Wang
- Department of UrologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jie Gao
- Department of NephrologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
7
|
Rahmoon MA, Elghaish RA, Ibrahim AA, Alaswad Z, Gad MZ, El-Khamisy SF, Elserafy M. High Glucose Increases DNA Damage and Elevates the Expression of Multiple DDR Genes. Genes (Basel) 2023; 14:144. [PMID: 36672885 PMCID: PMC9858638 DOI: 10.3390/genes14010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/19/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
The DNA Damage Response (DDR) pathways sense DNA damage and coordinate robust DNA repair and bypass mechanisms. A series of repair proteins are recruited depending on the type of breaks and lesions to ensure overall survival. An increase in glucose levels was shown to induce genome instability, yet the links between DDR and glucose are still not well investigated. In this study, we aimed to identify dysregulation in the transcriptome of normal and cancerous breast cell lines upon changing glucose levels. We first performed bioinformatics analysis using a microarray dataset containing the triple-negative breast cancer (TNBC) MDA-MB-231 and the normal human mammary epithelium MCF10A cell lines grown in high glucose (HG) or in the presence of the glycolysis inhibitor 2-deoxyglucose (2DG). Interestingly, multiple DDR genes were significantly upregulated in both cell lines grown in HG. In the wet lab, we remarkably found that HG results in severe DNA damage to TNBC cells as observed using the comet assay. In addition, several DDR genes were confirmed to be upregulated using qPCR analysis in the same cell line. Our results propose a strong need for DDR pathways in the presence of HG to oppose the severe DNA damage induced in cells.
Collapse
Affiliation(s)
- Mai A. Rahmoon
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza 12578, Egypt
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Reem A. Elghaish
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza 12578, Egypt
- University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Aya A. Ibrahim
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza 12578, Egypt
- University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Zina Alaswad
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza 12578, Egypt
- University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Mohamed Z. Gad
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Sherif F. El-Khamisy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza 12578, Egypt
- The Healthy Lifespan Institute and Institute of Neuroscience, School of Bioscience, University of Sheffield, Sheffield S10 2TN, UK
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1 DP, UK
| | - Menattallah Elserafy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza 12578, Egypt
- University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt
| |
Collapse
|
8
|
Bahloul A, Benayahoum A, Bouakkaz S, Bordjiba T, Boudjahem A, Lilya B, Bachari K. The antioxidant activity of N-E-caffeoyl and N-E-feruloyl tyramine conformers and their sulfured analogs contribution: density functional theory studies. Theor Chem Acc 2023. [DOI: 10.1007/s00214-022-02939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
9
|
Stancill JS, Corbett JA. Hydrogen peroxide detoxification through the peroxiredoxin/thioredoxin antioxidant system: A look at the pancreatic β-cell oxidant defense. VITAMINS AND HORMONES 2022; 121:45-66. [PMID: 36707143 PMCID: PMC10058777 DOI: 10.1016/bs.vh.2022.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Reactive oxygen species (ROS), such as hydrogen peroxide, are formed when molecular oxygen obtains additional electrons, increasing its reactivity. While low concentrations of hydrogen peroxide are necessary for regulation of normal cellular signaling events, high concentrations can be toxic. To maintain this balance between beneficial and deleterious concentrations of hydrogen peroxide, cells utilize antioxidants. Our recent work supports a primary role for peroxiredoxin, thioredoxin, and thioredoxin reductase as the oxidant defense pathway used by insulin-producing pancreatic β-cells. These three players work in an antioxidant cycle based on disulfide exchange, with oxidized targets ultimately being reduced using electrons provided by NADPH. Peroxiredoxins also participate in hydrogen peroxide-based signaling through disulfide exchange with redox-regulated target proteins. This chapter will describe the catalytic mechanisms of thioredoxin, thioredoxin reductase, and peroxiredoxin and provide an in-depth look at the roles these enzymes play in antioxidant defense pathways of insulin-secreting β-cells. Finally, we will evaluate the physiological relevance of peroxiredoxin-mediated hydrogen peroxide signaling as a regulator of β-cell function.
Collapse
Affiliation(s)
- Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
10
|
Stancill JS, Hansen PA, Mathison AJ, Schmidt EE, Corbett JA. Deletion of Thioredoxin Reductase Disrupts Redox Homeostasis and Impairs β-Cell Function. FUNCTION (OXFORD, ENGLAND) 2022; 3:zqac034. [PMID: 35873655 PMCID: PMC9301323 DOI: 10.1093/function/zqac034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 01/07/2023]
Abstract
Reactive oxygen species (ROS) have been implicated as mediators of pancreatic β-cell damage. While β-cells are thought to be vulnerable to oxidative damage, we have shown, using inhibitors and acute depletion, that thioredoxin reductase, thioredoxin, and peroxiredoxins are the primary mediators of antioxidant defense in β-cells. However, the role of this antioxidant cycle in maintaining redox homeostasis and β-cell survival in vivo remains unclear. Here, we generated mice with a β-cell specific knockout of thioredoxin reductase 1 (Txnrd1fl/fl; Ins1Cre/+ , βKO). Despite blunted glucose-stimulated insulin secretion, knockout mice maintain normal whole-body glucose homeostasis. Unlike pancreatic islets with acute Txnrd1 inhibition, βKO islets do not demonstrate increased sensitivity to ROS. RNA-sequencing analysis revealed that Txnrd1-deficient β-cells have increased expression of nuclear factor erythroid 2-related factor 2 (Nrf2)-regulated genes, and altered expression of genes involved in heme and glutathione metabolism, suggesting an adaptive response. Txnrd1-deficient β-cells also have decreased expression of factors controlling β-cell function and identity which may explain the mild functional impairment. Together, these results suggest that Txnrd1-knockout β-cells compensate for loss of this essential antioxidant pathway by increasing expression of Nrf2-regulated antioxidant genes, allowing for protection from excess ROS at the expense of normal β-cell function and identity.
Collapse
Affiliation(s)
| | - Polly A Hansen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226, USA
| | - Angela J Mathison
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA,Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Edward E Schmidt
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MN 59717, USA,Redox Biology Laboratory, University of Veterinary Medicine, Budapest 1078, Hungary
| | | |
Collapse
|
11
|
Dinić S, Arambašić Jovanović J, Uskoković A, Mihailović M, Grdović N, Tolić A, Rajić J, Đorđević M, Vidaković M. Oxidative stress-mediated beta cell death and dysfunction as a target for diabetes management. Front Endocrinol (Lausanne) 2022; 13:1006376. [PMID: 36246880 PMCID: PMC9554708 DOI: 10.3389/fendo.2022.1006376] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/05/2022] [Indexed: 11/14/2022] Open
Abstract
The biggest drawback of a current diabetes therapy is the treatment of the consequences not the cause of the disease. Regardless of the diabetes type, preservation and recovery of functional pancreatic beta cells stands as the biggest challenge in the treatment of diabetes. Free radicals and oxidative stress are among the major mediators of autoimmune destruction of beta cells in type 1 diabetes (T1D) or beta cell malfunction and death provoked by glucotoxicity and insulin resistance in type 2 diabetes (T2D). Additionally, oxidative stress reduces functionality of beta cells in T2D by stimulating their de-/trans-differentiation through the loss of transcription factors critical for beta cell development, maturity and regeneration. This review summarizes up to date clarified redox-related mechanisms involved in regulating beta cell identity and death, underlining similarities and differences between T1D and T2D. The protective effects of natural antioxidants on the oxidative stress-induced beta cell failure were also discussed. Considering that oxidative stress affects epigenetic regulatory mechanisms involved in the regulation of pancreatic beta cell survival and insulin secretion, this review highlighted huge potential of epigenetic therapy. Special attention was paid on application of the state-of-the-art CRISPR/Cas9 technology, based on targeted epigenome editing with the purpose of changing the differentiation state of different cell types, making them insulin-producing with ability to attenuate diabetes. Clarification of the above-mentioned mechanisms could provide better insight into diabetes etiology and pathogenesis, which would allow development of novel, potentially more efficient therapeutic strategies for the prevention or reversion of beta cell loss.
Collapse
|
12
|
Barone E, Di Domenico F, Perluigi M, Butterfield DA. The interplay among oxidative stress, brain insulin resistance and AMPK dysfunction contribute to neurodegeneration in type 2 diabetes and Alzheimer disease. Free Radic Biol Med 2021; 176:16-33. [PMID: 34530075 PMCID: PMC8595768 DOI: 10.1016/j.freeradbiomed.2021.09.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly followed by vascular dementia. In addition to clinically diagnosed dementia, cognitive dysfunction has been reported in diabetic patients. Recent studies are now beginning to recognize type 2 diabetes mellitus (T2DM), characterized by chronic hyperglycemia and insulin resistance, as a risk factor for AD and other cognitive disorders. While studies on insulin action have remained traditionally in the domain of peripheral tissues, the detrimental effects of insulin resistance in the central nervous system on cognitive dysfunction are increasingly being reported in recent clinical and preclinical studies. Brain functions require continuous supply of glucose and oxygen and a tight regulation of metabolic processes. Loss of this metabolic regulation has been proposed to be a contributor to memory dysfunction associated with neurodegeneration. Within the above scenario, this review will focus on the interplay among oxidative stress (OS), insulin resistance and AMPK dysfunctions in the brain by highlighting how these neurotoxic events contribute to neurodegeneration. We provide an overview on the detrimental effects of OS on proteins regulating insulin signaling and how these alterations impact cell metabolic dysfunctions through AMPK dysregulation. Such processes, we assert, are critically involved in the molecular pathways that underlie AD.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
13
|
Halliwell B, Adhikary A, Dingfelder M, Dizdaroglu M. Hydroxyl radical is a significant player in oxidative DNA damage in vivo. Chem Soc Rev 2021; 50:8355-8360. [PMID: 34128512 PMCID: PMC8328964 DOI: 10.1039/d1cs00044f] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent publications have suggested that oxidative DNA damage mediated by hydroxyl radical (˙OH) is unimportant in vivo, and that carbonate anion radical (CO3˙-) plays the key role. We examine these claims and summarize the evidence that ˙OH does play a key role as an important member of the reactive oxygen species (ROS) in vivo.
Collapse
Affiliation(s)
- Barry Halliwell
- Department of Biochemistry, National University of Singapore, Centre for Life Sciences, #05-01A, 28 Medical Drive, 117456, Singapore.
| | | | | | | |
Collapse
|
14
|
Stancill JS, Corbett JA. The Role of Thioredoxin/Peroxiredoxin in the β-Cell Defense Against Oxidative Damage. Front Endocrinol (Lausanne) 2021; 12:718235. [PMID: 34557160 PMCID: PMC8453158 DOI: 10.3389/fendo.2021.718235] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/19/2021] [Indexed: 02/02/2023] Open
Abstract
Oxidative stress is hypothesized to play a role in pancreatic β-cell damage, potentially contributing to β-cell dysfunction and death in both type 1 and type 2 diabetes. Oxidative stress arises when naturally occurring reactive oxygen species (ROS) are produced at levels that overwhelm the antioxidant capacity of the cell. ROS, including superoxide and hydrogen peroxide, are primarily produced by electron leak during mitochondrial oxidative metabolism. Additionally, peroxynitrite, an oxidant generated by the reaction of superoxide and nitric oxide, may also cause β-cell damage during autoimmune destruction of these cells. β-cells are thought to be susceptible to oxidative damage based on reports that they express low levels of antioxidant enzymes compared to other tissues. Furthermore, markers of oxidative damage are observed in islets from diabetic rodent models and human patients. However, recent studies have demonstrated high expression of various isoforms of peroxiredoxins, thioredoxin, and thioredoxin reductase in β-cells and have provided experimental evidence supporting a role for these enzymes in promoting β-cell function and survival in response to a variety of oxidative stressors. This mini-review will focus on the mechanism by which thioredoxins and peroxiredoxins detoxify ROS and on the protective roles of these enzymes in β-cells. Additionally, we speculate about the role of this antioxidant system in promoting insulin secretion.
Collapse
|
15
|
Zheng YZ, Deng G, Guo R, Fu ZM, Chen DF. Effects of different ester chains on the antioxidant activity of caffeic acid. Bioorg Chem 2020; 105:104341. [DOI: 10.1016/j.bioorg.2020.104341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 11/29/2022]
|
16
|
Stancill JS, Happ JT, Broniowska KA, Hogg N, Corbett JA. Peroxiredoxin 1 plays a primary role in protecting pancreatic β-cells from hydrogen peroxide and peroxynitrite. Am J Physiol Regul Integr Comp Physiol 2020; 318:R1004-R1013. [PMID: 32292063 DOI: 10.1152/ajpregu.00011.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Both reactive nitrogen and oxygen species (RNS and ROS), such as nitric oxide, peroxynitrite, and hydrogen peroxide, have been implicated as mediators of pancreatic β-cell damage during the pathogenesis of autoimmune diabetes. While β-cells are thought to be vulnerable to oxidative damage due to reportedly low levels of antioxidant enzymes, such as catalase and glutathione peroxidase, we have shown that they use thioredoxin reductase to detoxify hydrogen peroxide. Thioredoxin reductase is an enzyme that participates in the peroxiredoxin antioxidant cycle. Peroxiredoxins are expressed in β-cells and, when overexpressed, protect against oxidative stress, but the endogenous roles of peroxiredoxins in the protection of β-cells from oxidative damage are unclear. Here, using either glucose oxidase or menadione to continuously deliver hydrogen peroxide, or the combination of dipropylenetriamine NONOate and menadione to continuously deliver peroxynitrite, we tested the hypothesis that β-cells use peroxiredoxins to detoxify both of these reactive species. Either pharmacological peroxiredoxin inhibition with conoidin A or specific depletion of cytoplasmic peroxiredoxin 1 (Prdx1) using siRNAs sensitizes INS 832/13 cells and rat islets to DNA damage and death induced by hydrogen peroxide or peroxynitrite. Interestingly, depletion of peroxiredoxin 2 (Prdx2) had no effect. Together, these results suggest that β-cells use cytoplasmic Prdx1 as a primary defense mechanism against both ROS and RNS.
Collapse
Affiliation(s)
- Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John T Happ
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Neil Hogg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
17
|
Zheng YZ, Zhou Y, Guo R, Fu ZM, Chen DF. Structure-antioxidant activity relationship of ferulic acid derivatives: Effect of ester groups at the end of the carbon side chain. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2019.108932] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
18
|
Norouzirad R, Gholami H, Ghanbari M, Hedayati M, González-Muniesa P, Jeddi S, Ghasemi A. Dietary inorganic nitrate attenuates hyperoxia-induced oxidative stress in obese type 2 diabetic male rats. Life Sci 2019; 230:188-196. [PMID: 31150686 DOI: 10.1016/j.lfs.2019.05.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/18/2019] [Accepted: 05/26/2019] [Indexed: 10/26/2022]
Abstract
AIMS Hyperoxia has beneficial metabolic effects in type 2 diabetes. However, hyperoxia exacerbates already existing oxidative stress in type 2 diabetes. Nitrate, a nitric oxide donor, is an effective new treatment in type 2 diabetes and also has antioxidant properties. The aim of this study was to determine whether nitrate administration can attenuate hyperoxia-induced oxidative stress in obese type 2 diabetic rats. MAIN METHODS Fifty-six male Wistar rats (190-210 g) were divided into 8 groups: Controls (non-treated, nitrate-treated, O2-treated, and nitrate + O2-treated) and diabetes (non-treated, nitrate-treated, O2-treated, and nitrate + O2-treated). Diabetes was induced using high-fat diet and low-dose of streptozotocin (30 mg/kg). Rats in intervention groups, were exposed to 95% oxygen and consumed sodium nitrate (100 mg/L) in drinking water. Serum fasting glucose, oxidized (GSSG) and reduced (GSH) glutathiones, total oxidant status (TOS), catalase and superoxide dismutase (SOD) activities, and total antioxidant capacity (TAC) were measured after intervention. Oxidative stress index (OSI) was calculated as TOS/TAC ratio. KEY FINDINGS Diabetic rats had increased oxidative stress and hyperoxia exacerbated it. In O2-diabetic rats, nitrate decreased GSSG (102.7 ± 2.1 vs. 236.0 ± 20.1 μM, P < 0.001), TOS (67.7 ± 7.3 vs. 104 ± 3.8 μM, P < 0.001), and OSI (0.44 ± 0.04 vs. 0.91 ± 0.07, P < 0.001) and increased catalase (2.8 ± 0.13 vs. 1.8 ± 0.21 KU/L, P = 0.014), SOD (53.4 ± 1.5 vs. 38.4 ± 1.2 U/mL, P < 0.001), GSH (43.7 ± 1.4 vs. 17.8 ± 0.5 mM, P = 0.003), TAC (152.5 ± 1.9 vs. 116.7 ± 5.0 mM, P < 0.001), and GSH/GSSG ratio (0.43 ± 0.01 vs. 0.08 ± 0.01, P = 0.005). Nitrate also potentiated effects of hyperoxia on decreasing fasting glucose. SIGNIFICANCE Our results showed that dietary nitrate attenuates hyperoxia-induced oxidative stress in type 2 diabetic rats.
Collapse
Affiliation(s)
- Reza Norouzirad
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Dezful University of Medical Sciences, Dezful, Iran.
| | - Hanieh Gholami
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Ghanbari
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pedro González-Muniesa
- University of Navarra, Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, Pamplona, Spain; University of Navarra, Centre for Nutrition Research, School of Pharmacy and Nutrition, Pamplona, Spain; IdiSNA Navarra's Health Research Institute, Pamplona, Spain; CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain.
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Antioxidant activity and pKa calculations of 4‑mercaptostilbene and some derivatives: A theoretical approach. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2018.11.092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
20
|
Stancill JS, Broniowska KA, Oleson BJ, Naatz A, Corbett JA. Pancreatic β-cells detoxify H 2O 2 through the peroxiredoxin/thioredoxin antioxidant system. J Biol Chem 2019; 294:4843-4853. [PMID: 30659092 DOI: 10.1074/jbc.ra118.006219] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
Oxidative stress is thought to promote pancreatic β-cell dysfunction and contribute to both type 1 and type 2 diabetes. Reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, are mediators of oxidative stress that arise largely from electron leakage during oxidative phosphorylation. Reports that β-cells express low levels of antioxidant enzymes, including catalase and GSH peroxidases, have supported a model in which β-cells are ill-equipped to detoxify ROS. This hypothesis seems at odds with the essential role of β-cells in the control of metabolic homeostasis and organismal survival through exquisite coupling of oxidative phosphorylation, a prominent ROS-producing pathway, to insulin secretion. Using glucose oxidase to deliver H2O2 continuously over time and Amplex Red to measure extracellular H2O2 concentration, we found here that β-cells can remove micromolar levels of this oxidant. This detoxification pathway utilizes the peroxiredoxin/thioredoxin antioxidant system, as selective chemical inhibition or siRNA-mediated depletion of thioredoxin reductase sensitized β-cells to continuously generated H2O2 In contrast, when delivered as a bolus, H2O2 induced the DNA damage response, depleted cellular energy stores, and decreased β-cell viability independently of thioredoxin reductase inhibition. These findings show that β-cells have the capacity to detoxify micromolar levels of H2O2 through a thioredoxin reductase-dependent mechanism and are not as sensitive to oxidative damage as previously thought.
Collapse
Affiliation(s)
- Jennifer S Stancill
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Katarzyna A Broniowska
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Bryndon J Oleson
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Aaron Naatz
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - John A Corbett
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
21
|
Choi SW, Ho CK. Antioxidant properties of drugs used in Type 2 diabetes management: could they contribute to, confound or conceal effects of antioxidant therapy? Redox Rep 2018; 23:1-24. [PMID: 28514939 PMCID: PMC6748682 DOI: 10.1080/13510002.2017.1324381] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES This is a narrative review, investigating the antioxidant properties of drugs used in the management of diabetes, and discusses whether these antioxidant effects contribute to, confound, or conceal the effects of antioxidant therapy. METHODS A systematic search for articles reporting trials, or observational studies on the antioxidant effect of drugs used in the treatment of diabetes in humans or animals was performed using Web of Science, PubMed, and Ovid. Data were extracted, including data on a number of subjects, type of treatment (and duration) received, and primary and secondary outcomes. The primary outcomes were reporting on changes in biomarkers of antioxidants concentrations and secondary outcomes were reporting on changes in biomarkers of oxidative stress. RESULTS Diabetes Mellitus is a disease characterized by increased oxidative stress. It is often accompanied by a spectrum of other metabolic disturbances, including elevated plasma lipids, elevated uric acid, hypertension, endothelial dysfunction, and central obesity. This review shows evidence that some of the drugs in diabetes management have both in vivo and in vitro antioxidant properties through mechanisms such as scavenging free radicals and upregulating antioxidant gene expression. CONCLUSION Pharmaceutical agents used in the treatment of type 2 diabetes has been shown to exert an antioxidant effect..
Collapse
Affiliation(s)
- Siu Wai Choi
- Department of Anesthesiology, Queen Mary
Hospital, The University of Hong Kong, Pokfulam, Hong
Kong SAR
| | - Cyrus K. Ho
- Faculty of Veterinary and Agricultural
Sciences, The University of Melbourne, Melbourne,
Australia
- Faculty of Health and Social Sciences,
School of Nursing, The Hong Kong Polytechnic University,
Kowloon, Hong Kong SAR
| |
Collapse
|
22
|
Evans LW, Omaye ST. Use of Saliva Biomarkers to Monitor Efficacy of Vitamin C in Exercise-Induced Oxidative Stress. Antioxidants (Basel) 2017; 6:E5. [PMID: 28085082 PMCID: PMC5384169 DOI: 10.3390/antiox6010005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/19/2016] [Accepted: 01/09/2017] [Indexed: 02/07/2023] Open
Abstract
Saliva is easily obtainable for medical research and requires little effort or training for collection. Because saliva contains a variety of biological compounds, including vitamin C, malondialdehyde, amylase, and proteomes, it has been successfully used as a biospecimen for the reflection of health status. A popular topic of discussion in medical research is the potential association between oxidative stress and negative outcomes. Systemic biomarkers that represent oxidative stress can be found in saliva. It is unclear, however, if saliva is an accurate biospecimen as is blood and/or plasma. Exercise can induce oxidative stress, resulting in a trend of antioxidant supplementation to combat its assumed detriments. Vitamin C is a popular antioxidant supplement in the realm of sports and exercise. One potential avenue for evaluating exercise induced oxidative stress is through assessment of biomarkers like vitamin C and malondialdehyde in saliva. At present, limited research has been done in this area. The current state of research involving exercise-induced oxidative stress, salivary biomarkers, and vitamin C supplementation is reviewed in this article.
Collapse
Affiliation(s)
- Levi W Evans
- Nutrition Program, Agriculture, Nutrition and Veterinary Science Department, University of Nevada, Reno, NV 89557, USA.
| | - Stanley T Omaye
- Nutrition Program, Agriculture, Nutrition and Veterinary Science Department, University of Nevada, Reno, NV 89557, USA.
| |
Collapse
|
23
|
Nasif WA, Mukhtar MH, Nour Eldein MM, Ashgar SS. Oxidative DNA damage and oxidized low density lipoprotein in Type II diabetes mellitus among patients with Helicobacter pylori infection. Diabetol Metab Syndr 2016; 8:34. [PMID: 27148410 PMCID: PMC4855822 DOI: 10.1186/s13098-016-0149-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection is reported to be associated with various extragastrointestinal conditions such as insulin resistance, diabetes mellitus and metabolic syndrome. H. pylori infection and type 2 diabetes mellitus (T2DM) are associated with oxidative stress, this cross-relation between H. pylori induced infection in T2DM and oxidative damage is still debated. Thus, the question arises whether an increase in the serum level of 8-OHdG and Ox-LDL will occurs in patients with T2DM infected H. pylori; this will be through determination and compare frequency of H. pylori infection in T2DM and non-diabetic patients. METHODS 100 patients presented with history of epigastric discomfort for more than 1 month; 50 patients with T2DM and 50 non-diabetics. Anti-H. pylori IgG using ELISA, fasting and postprandial glucose level, glycated hemoglobin (HbA1c) and body mass index (BMI) was calculated. Serum 8-OHdG and Ox-LDL was measured using ELISA for the 100 patients and 50 control subject. RESULTS Rates of H. pylori infection of T2DM and non-diabetic were 66 and 58 %, respectively, (p = 0.001). H. pylori IgG antibody was not correlated with HbA1c either in T2DM (p = 0.06) or non-diabetic (p = 0.25). Serum 8-OHdG level in T2DM with positive H. pylori infection showed a significant difference compared to non-diabetics with positive H. pylori infection (p = 0.001) and higher than that in T2DM with negative H. pylori. A correlation between 8-OHdG concentration and HbA1c in T2DM patients infected with H. pylori was observed (r = 0.39, p = 0.02). Serum Ox-LDL level in T2DM with positive H. pylori infection showed a significant difference compared to diabetics with both negative H. pylori infection and in non-diabetics with positive H. pylori infection (p = 0.001). CONCLUSIONS Increased levels of oxidative DNA damage (8-OHdG) and Ox-LDL suggest the mechanistic link between H. pylori infection combined with diabetes and increased generation of ROS and could play as an important image for high risk to atherosclerosis.
Collapse
Affiliation(s)
- Wesam Ahmed Nasif
- />Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
- />Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Sadat City, Egypt
| | - Mohammed Hasan Mukhtar
- />Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
| | - Mohammed Mahmoud Nour Eldein
- />Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
- />Oncology Diagnostic Unit, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sami Sadagah Ashgar
- />Microbiology Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
| |
Collapse
|
24
|
Diverse effects of naturally occurring base lesions on the structure and stability of the human telomere DNA quadruplex. Biochimie 2015; 118:15-25. [DOI: 10.1016/j.biochi.2015.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/13/2015] [Indexed: 12/28/2022]
|
25
|
Rouaiguia-Bouakkaz S, Benayahoum A. The antioxidant activity of 4-hydroxycoumarin derivatives and some sulfured analogs. J PHYS ORG CHEM 2015. [DOI: 10.1002/poc.3475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Ali Benayahoum
- Laboratory of Applied Chemistry, 8 Mai 1945; Guelma University; Guelma 24000 Algeria
- Research Center in Analytical Chemistry and Physics (CRAPC); BP 248, Algiers RP Algiers 16004 Algeria
| |
Collapse
|
26
|
Blank ID, Sadeghian K, Ochsenfeld C. A base-independent repair mechanism for DNA glycosylase--no discrimination within the active site. Sci Rep 2015; 5:10369. [PMID: 26013033 PMCID: PMC4445063 DOI: 10.1038/srep10369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/09/2015] [Indexed: 11/09/2022] Open
Abstract
The ubiquitous occurrence of DNA damages renders its repair machinery a crucial requirement for the genomic stability and the survival of living organisms. Deficiencies in DNA repair can lead to carcinogenesis, Alzheimer, or Diabetes II, where increased amounts of oxidized DNA bases have been found in patients. Despite the highest mutation frequency among oxidized DNA bases, the base-excision repair process of oxidized and ring-opened guanine, FapydG (2,6-diamino-4-hydroxy-5-formamidopyrimidine), remained unclear since it is difficult to study experimentally. We use newly-developed linear-scaling quantum-chemical methods (QM) allowing us to include up to 700 QM-atoms and achieving size convergence. Instead of the widely assumed base-protonated pathway we find a ribose-protonated repair mechanism which explains experimental observations and shows strong evidence for a base-independent repair process. Our results also imply that discrimination must occur during recognition, prior to the binding within the active site.
Collapse
Affiliation(s)
- Iris D Blank
- 1] Chair of Theoretical Chemistry, Department of Chemistry, University of Munich (LMU), Butenandtstr. 7, D-81377 Munich, Germany [2] Center for Integrated Protein Science Munich (CIPSM) at the Department of Chemistry, University of Munich (LMU), Butenandtstr. 5-13, D-81377 Munich, Germany
| | - Keyarash Sadeghian
- 1] Chair of Theoretical Chemistry, Department of Chemistry, University of Munich (LMU), Butenandtstr. 7, D-81377 Munich, Germany [2] Center for Integrated Protein Science Munich (CIPSM) at the Department of Chemistry, University of Munich (LMU), Butenandtstr. 5-13, D-81377 Munich, Germany
| | - Christian Ochsenfeld
- 1] Chair of Theoretical Chemistry, Department of Chemistry, University of Munich (LMU), Butenandtstr. 7, D-81377 Munich, Germany [2] Center for Integrated Protein Science Munich (CIPSM) at the Department of Chemistry, University of Munich (LMU), Butenandtstr. 5-13, D-81377 Munich, Germany
| |
Collapse
|
27
|
Oxidative DNA damage is involved in cigarette smoke-induced lung injury in rats. Environ Health Prev Med 2015; 20:318-24. [PMID: 25967734 DOI: 10.1007/s12199-015-0469-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/30/2015] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE Reactive oxygen species (ROS) induced by exogenous toxicants are suggested to be involved in carcinogenesis by oxidative modification of DNA. 8-Hydroxyl-2-deoxyguanosine (8-OHdG) has been considered as a reliable biomarker for oxidative DNA damage both in vivo and in vitro studies. But the effect of smoking on oxidative damage has not yet been fully elucidated. METHODS Wistar rats were exposed to cigarette smoke at concentrations of 20 and 60 % for 30 min, twice/day for 45 weeks. Then the histopathology of lung tissues, levels of ROS, 8-OHdG, and total antioxidant (T-AOC), expression of DNA repair enzymes, e.g. 8-oxyguaine DNA glycosylase (OGG1), and MutThomolog 1 (Oxidized Purine Nucleoside Triphosphatase, MTH1) were determined in urine, peripheral blood lymphocytes, and lung tissue. RESULTS The results showed that long-term cigarette smoke exposure can cause obvious damages of lung tissue in rats. In addition, a significant and cigarette smoke concentration-dependent increase in ROS and 8-OHdG were observed compared with the non-exposed control rats. In contrast, the expression of OGG1 and MTH1, and T-AOC levels were obviously decreased after long-term exposure to cigarette smoke. CONCLUSION These findings indicate that long-term exposure to cigarette smoker increases ROS levels, decreases total antioxidant capacity, and interferes DNA repair capacity that eventually induces oxidative DNA damage, which appears to play an important role in cigarette smoke-induced lung injury in rats, and determination of 8-OHdG levels might be a useful method for monitoring oxidative damage in cigarette smokers.
Collapse
|
28
|
Karaman A, Aydın H, Geçkinli B, Çetinkaya A, Karaman S. DNA damage is increased in lymphocytes of patients with metabolic syndrome. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 782:30-5. [DOI: 10.1016/j.mrgentox.2015.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/07/2015] [Accepted: 03/10/2015] [Indexed: 10/23/2022]
|
29
|
Jones DA, Prior SL, Barry JD, Caplin S, Baxter JN, Stephens JW. Changes in markers of oxidative stress and DNA damage in human visceral adipose tissue from subjects with obesity and type 2 diabetes. Diabetes Res Clin Pract 2014; 106:627-33. [PMID: 25458337 DOI: 10.1016/j.diabres.2014.09.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/24/2014] [Accepted: 09/14/2014] [Indexed: 01/18/2023]
Abstract
AIMS In the past 30 years, prevalence of obesity has almost trebled resulting in an increased incidence of type 2 diabetes mellitus and other co-morbidities. Visceral adipose tissue is believed to play a vital role, but underlying mechanisms remain unclear. Our aim was to investigate changes in markers of oxidative damage in human visceral adipose tissue to determine levels of oxidative burden that may be attributed to obesity and/or diabetes. METHODS Visceral adipose tissue samples from 61 subjects undergoing abdominal surgery grouped as lean, obese and obese with type 2 diabetes mellitus, were examined using 3 different markers of oxidative stress. Malondialdehyde (MDA) concentration was measured as a marker of lipid peroxidation, telomere length and Comet assay as markers of oxidative DNA damage. RESULTS No significant difference in MDA concentration, telomere length and DNA damage was observed between groups, although longer telomere lengths were seen in the obese with diabetes group compared to the obese group (P<0.05). Lower MDA concentration and longer telomere length were seen in subjects with diabetes compared to those without (P<0.05). DNA damage, analysed via Comet assay, was significantly lower in subjects with diabetes compared to those without (P<0.05). CONCLUSION A paradoxical decrease in oxidative stress and DNA damage was observed in samples from subjects with type 2 diabetes mellitus. Further work is required to investigate this further, however this phenomenon may be due to an up regulation of antioxidant defences in adipose tissue.
Collapse
Affiliation(s)
- D A Jones
- Diabetes Research Group, College of Medicine, Swansea University, Singleton Park, Swansea, UK.
| | - S L Prior
- Diabetes Research Group, College of Medicine, Swansea University, Singleton Park, Swansea, UK
| | - J D Barry
- General Surgery (Upper GI (Obesity) and Endocrinology), Morriston Hospital, Swansea, UK
| | - S Caplin
- General Surgery (Upper GI (Obesity) and Endocrinology), Morriston Hospital, Swansea, UK
| | - J N Baxter
- General Surgery (Upper GI (Obesity) and Endocrinology), Morriston Hospital, Swansea, UK
| | - J W Stephens
- Diabetes Research Group, College of Medicine, Swansea University, Singleton Park, Swansea, UK; General Surgery (Upper GI (Obesity) and Endocrinology), Morriston Hospital, Swansea, UK
| |
Collapse
|
30
|
Huang Y, Cai X, Qiu M, Chen P, Tang H, Hu Y, Huang Y. Prediabetes and the risk of cancer: a meta-analysis. Diabetologia 2014; 57:2261-2269. [PMID: 25208757 DOI: 10.1007/s00125-014-3361-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/31/2014] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS The results from prospective cohort studies of prediabetes (impaired fasting glucose and/or impaired glucose tolerance) and risk of cancer are controversial. We conducted a meta-analysis to evaluate the risk of cancer in association with impaired fasting glucose and impaired glucose tolerance. METHODS The PubMed, EMBASE and Cochrane Library databases were searched for prospective cohort studies with data on prediabetes and cancer. Two independent reviewers assessed the reports and extracted the data. Prospective studies were included if they reported adjusted RRs with 95% CIs for the association between cancer and prediabetes. Subgroup analyses were conducted according to endpoint, age, sex, ethnicity, duration of follow-up and study characteristics. RESULTS Data from 891,426 participants were derived from 16 prospective cohort studies. Prediabetes was associated with an increased risk of cancer overall (RR 1.15; 95% CI 1.06, 1.23). The results were consistent across cancer endpoint, age, duration of follow-up and ethnicity. There was no significant difference for the risk of cancer with different definitions of prediabetes. In a site-specific cancer analysis, prediabetes was significantly associated with increased risks of cancer of the stomach/colorectum, liver, pancreas, breast and endometrium (all p < 0.05), but not associated with cancer of the bronchus/lung, prostate, ovary, kidney or bladder. The risks of site-specific cancer were significantly different (p = 0.01) and were highest for liver, endometrial and stomach/colorectal cancer. CONCLUSIONS/INTERPRETATION Overall, prediabetes was associated with an increased risk of cancer, especially liver, endometrial and stomach/colorectal cancer.
Collapse
Affiliation(s)
- Yi Huang
- Clinical Medicine Research Center, The First People's Hospital of Shunde, Penglai Road, Daliang Town, Shunde District, Foshan, 528300, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
31
|
Kang L, Dai C, Lustig ME, Bonner JS, Mayes WH, Mokshagundam S, James FD, Thompson CS, Lin CT, Perry CGR, Anderson EJ, Neufer PD, Wasserman DH, Powers AC. Heterozygous SOD2 deletion impairs glucose-stimulated insulin secretion, but not insulin action, in high-fat-fed mice. Diabetes 2014; 63:3699-710. [PMID: 24947366 PMCID: PMC4207395 DOI: 10.2337/db13-1845] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Elevated reactive oxygen species (ROS) are linked to insulin resistance and islet dysfunction. Manganese superoxide dismutase (SOD2) is a primary defense against mitochondrial oxidative stress. To test the hypothesis that heterozygous SOD2 deletion impairs glucose-stimulated insulin secretion (GSIS) and insulin action, wild-type (sod2(+/+)) and heterozygous knockout mice (sod2(+/-)) were fed a chow or high-fat (HF) diet, which accelerates ROS production. Hyperglycemic (HG) and hyperinsulinemic-euglycemic (HI) clamps were performed to assess GSIS and insulin action in vivo. GSIS during HG clamps was equal in chow-fed sod2(+/-) and sod2(+/+) but was markedly decreased in HF-fed sod2(+/-). Remarkably, this impairment was not paralleled by reduced HG glucose infusion rate (GIR). Decreased GSIS in HF-fed sod2(+/-) was associated with increased ROS, such as superoxide ion. Surprisingly, insulin action determined by HI clamps did not differ between sod2(+/-) and sod2(+/+) of either diet. Since insulin action was unaffected, we hypothesized that the unchanged HG GIR in HF-fed sod2(+/-) was due to increased glucose effectiveness. Increased GLUT-1, hexokinase II, and phospho-AMPK protein in muscle of HF-fed sod2(+/-) support this hypothesis. We conclude that heterozygous SOD2 deletion in mice, a model that mimics SOD2 changes observed in diabetic humans, impairs GSIS in HF-fed mice without affecting insulin action.
Collapse
Affiliation(s)
- Li Kang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN Division of Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, U.K.
| | - Chunhua Dai
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, TN
| | - Mary E Lustig
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Jeffrey S Bonner
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Wesley H Mayes
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Shilpa Mokshagundam
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Freyja D James
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Courtney S Thompson
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, TN
| | - Chien-Te Lin
- East Carolina Diabetes and Obesity Institute and Departments of Physiology and Kinesiology, East Carolina University, Greenville, NC
| | - Christopher G R Perry
- East Carolina Diabetes and Obesity Institute and Departments of Physiology and Kinesiology, East Carolina University, Greenville, NC
| | - Ethan J Anderson
- East Carolina Diabetes and Obesity Institute and Departments of Physiology and Kinesiology, East Carolina University, Greenville, NC
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute and Departments of Physiology and Kinesiology, East Carolina University, Greenville, NC
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, TN Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
| |
Collapse
|
32
|
Raj PS, Prathapan A, Sebastian J, Antony AK, Riya MP, Rani MRP, Biju H, Priya S, Raghu KG. Parmotrema tinctorum exhibits antioxidant, antiglycation and inhibitory activities against aldose reductase and carbohydrate digestive enzymes: an in vitro study. Nat Prod Res 2014; 28:1480-4. [PMID: 24735436 DOI: 10.1080/14786419.2014.909420] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study evaluated the inhibitory potential of ethyl acetate extract of Parmotrema tinctorum (PTEE), an edible lichen, against aldose reductase (AR) and carbohydrate digestive enzymes such as α-glucosidase and α-amylase. It was also screened for antioxidant activities by using DPPH, ABTS, superoxide and hydroxyl radical-scavenging assays. PTEE exhibited α-glucosidase, α-amylase and AR inhibition along with significant antiglycation potential with an estimated IC50 value of 58.45 ± 1.24, 587.74 ± 3.27, 139.28 ± 2.6 and 285.78 ± 1.287 μg/mL, respectively. Antioxidant activity of PTEE against DPPH (IC50 396.83 ± 2.98 μg/mL), ABTS (151.34 ± 1.79 μg/mL), superoxide (30.29 ± 1.17 μg/mL) and hydroxyl (35.42 ± 1.22 μg/mL) radicals suggests the antioxidant potential of P. tinctorum. Significant antioxidant activity and inhibitory potential against carbohydrate digestive enzymes and AR suggest that P. tinctorum can be developed as functional food/nutraceuticals for diabetes after detailed study.
Collapse
Affiliation(s)
- P Salin Raj
- a Agroprocessing and Natural Products Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST) , Pappanamcode, Thiruvananthapuram , Kerala 695019 , India
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ishiki M, Nishida Y, Ishibashi H, Wada T, Fujisaka S, Takikawa A, Urakaze M, Sasaoka T, Usui I, Tobe K. Impact of divergent effects of astaxanthin on insulin signaling in L6 cells. Endocrinology 2013; 154:2600-12. [PMID: 23715867 DOI: 10.1210/en.2012-2198] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Because oxidative stress promotes insulin resistance in obesity and type 2 diabetes, it is crucial to find effective antioxidant for the purpose of decreasing this threat. In this study, we explored the effect of astaxanthin, a carotenoid antioxidant, on insulin signaling and investigated whether astaxanthin improves cytokine- and free fatty acid-induced insulin resistance in vitro. We examined the effect of astaxanthin on insulin-stimulated glucose transporter 4 (GLUT4) translocation, glucose uptake, and insulin signaling in cultured rat L6 muscle cells using plasma membrane lawn assay, 2-deoxyglucose uptake, and Western blot analysis. Next, we examined the effect of astaxanthin on TNFα- and palmitate-induced insulin resistance. The amount of reactive oxygen species generated by TNFα or palmitate with or without astaxanthin was evaluated by dichlorofluorescein staining. We also compared the effect of astaxanthin on insulin signaling with that of other antioxidants, α-lipoic acid and α-tocopherol. We observed astaxanthin enhanced insulin-stimulated GLUT4 translocation and glucose uptake, which was associated with an increase in insulin receptor substrate-1 tyrosine and Akt phosphorylation and a decrease in c-Jun N-terminal kinase (JNK) and insulin receptor substrate-1 serine 307 phosphorylation. Furthermore, astaxanthin restored TNFα- and palmitate-induced decreases in insulin-stimulated GLUT4 translocation or glucose uptake with a concomitant decrease in reactive oxygen species generation. α-Lipoic acid enhanced Akt phosphorylation and decreased ERK and JNK phosphorylation, whereas α-tocopherol enhanced ERK and JNK phosphorylation but had little effect on Akt phosphorylation. Collectively these findings indicate astaxanthin is a very effective antioxidant for ameliorating insulin resistance by protecting cells from oxidative stress generated by various stimuli including TNFα and palmitate.
Collapse
Affiliation(s)
- Manabu Ishiki
- The First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Karunakaran U, Park KG. A systematic review of oxidative stress and safety of antioxidants in diabetes: focus on islets and their defense. Diabetes Metab J 2013; 37:106-12. [PMID: 23641350 PMCID: PMC3638220 DOI: 10.4093/dmj.2013.37.2.106] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
A growing body of evidence suggests that hyperglycemia-induced oxidative stress plays an important role in diabetic complications, especially β-cell dysfunction and failure. Under physiological conditions, reactive oxygen species serve as second messengers that facilitate signal transduction and gene expression in pancreatic β-cells. However, under pathological conditions, an imbalance in redox homeostasis leads to aberrant tissue damage and β-cell death due to a lack of antioxidant defense systems. Taking into account the vulnerability of islets to oxidative damage, induction of endogenous antioxidant enzymes or exogenous antioxidant administration has been proposed as a way to protect β-cells against diabetic insults. Here, we consider recent insights into how the redox response becomes deregulated under diabetic conditions, as well as the therapeutic benefits of antioxidants, which may provide clues for developing strategies aimed at the treatment or prevention of diabetes associated with β-cell failure.
Collapse
Affiliation(s)
- Udayakumar Karunakaran
- Departments of Internal Medicine, Biochemistry and Cell Biology, Research Institute of Aging and Metabolism and World Class University Program, Kyungpook National University School of Medicine, Daegu, Korea
| | - Keun-Gyu Park
- Departments of Internal Medicine, Biochemistry and Cell Biology, Research Institute of Aging and Metabolism and World Class University Program, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
35
|
Prathapan A, Krishna MS, Nisha V, Sundaresan A, Raghu K. Polyphenol rich fruit pulp of Aegle marmelos (L.) Correa exhibits nutraceutical properties to down regulate diabetic complications — An in vitro study. Food Res Int 2012. [DOI: 10.1016/j.foodres.2012.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Nie JH, Chen ZH, Liu X, Wu YW, Li JX, Cao Y, Hei TK, Tong J. Oxidative damage in various tissues of rats exposed to radon. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2012; 75:694-9. [PMID: 22757673 DOI: 10.1080/15287394.2012.690086] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Oxidative damage can be induced by many environmental stressors. 8-Hydroxydeoxyguanosine (8-OHdG) has been used as a biomarker of oxidative DNA damage in both in vitro and in vivo studies. In the present study, Wistar rats were exposed to radon gas at a concentration of 100,000Bq/m(3) for 12 h/d for 30, 60, and 120 d, equivalent to cumulative doses of 60, 120, and 240 working level months (WLM), respectively. Changes in levels of 8-OHdG, reactive oxygen species (ROS), and total antioxidant (T-AOC), as well as expressions of some DNA repair enzymes such as 8-oxoguanine DNA glycosylase (OGG1) and MutT homolog 1 (oxidized purine nucleoside triphosphatase, MTH1), were determined in rat urine, peripheral blood lymphocytes, and lung after exposure to radon. The results revealed an increase in 8-OHdG and ROS levels, a decrease in T-AOC levels, and reduced OGG1 and MTH1 expression levels. The elevated amount of 8-OHdG in urine or lymphocytes was positively correlated with the cumulative exposure dose, whereas OGG1 and MHT1 expression levels in lung were inversely correlated with cumulative exposure dose. These findings indicate that oxidative damage induced by radon may be involved in radon-induced carcinogenesis.
Collapse
Affiliation(s)
- Ji-Hua Nie
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Rabbani SI, Devi K, Khanam S. Protective role of glibenclamide against nicotinamide-streptozotocin induced nuclear damage in diabetic Wistar rats. J Pharmacol Pharmacother 2011; 1:18-23. [PMID: 21808586 PMCID: PMC3142752 DOI: 10.4103/0976-500x.64531] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Objective: To evaluate the protective effect of glibenclamide against the experimental diabetes-induced nuclear damage in Wistar rats. Materials and Methods: The anti-mutagenic effect of glibenclamide (0.5, 5 and 50 mg/kg, p.o daily for 4 weeks) was evaluated against the nicotinamide (NA)-streptozotocin (STZ) induced type-2 diabetes mellitus using bone marrow micronucleus and sperm abnormalities tests. The antioxidant status was tested by estimating the serum levels of lipid peroxidation (LPO), catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Results: The results indicated that glibenclamide at 50 mg/kg decreased the frequency of micronuclei in erythrocytes (P < 0.05) and sperm shape abnormality (P < 0.01) besides enhancing the antioxidant status (P < 0.05) in the diabetic rats. However, glibenclamide treatment did not enhance the polychromatic and normochromatic erythrocytes (P/N) ratio and sperm count in the diabetic condition. Conclusion: The observations indicate that the glibenclamide has anti-mutagenic potential which could be related to the antioxidant effect and might also possess anti-proliferative property.
Collapse
Affiliation(s)
- Syed Imam Rabbani
- Department of Pharmacology, Al-Ameen College of Pharmacy, Opp. Lalbagh Main Gate, Hosur Road, Bangalore-560 027, India
| | | | | |
Collapse
|
38
|
Ovadia H, Haim Y, Nov O, Almog O, Kovsan J, Bashan N, Benhar M, Rudich A. Increased adipocyte S-nitrosylation targets anti-lipolytic action of insulin: relevance to adipose tissue dysfunction in obesity. J Biol Chem 2011; 286:30433-30443. [PMID: 21724851 DOI: 10.1074/jbc.m111.235945] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Protein S-nitrosylation is a reversible protein modification implicated in both physiological and pathophysiological regulation of protein function. In obesity, skeletal muscle insulin resistance is associated with increased S-nitrosylation of insulin-signaling proteins. However, whether adipose tissue is similarly affected in obesity and, if so, what are the causes and functional consequences of increased S-nitrosylation in this tissue are unknown. Total protein S-nitrosylation was increased in intra-abdominal adipose tissue of obese humans and in high fat-fed or leptin-deficient ob/ob mice. Both the insulin receptor β-subunit and Akt were S-nitrosylated, correlating with body weight. Elevated protein and mRNA expression of inducible NO synthase and decreased protein levels of thioredoxin reductase were associated with increased adipose tissue S-nitrosylation. Cultured differentiated pre-adipocyte cell lines exposed to the NO donors S-nitrosoglutathione (GSNO) or S-nitroso-N-acetylpenicillamine exhibited diminished insulin-stimulated phosphorylation of Akt but not of GSK3 nor of insulin-stimulated glucose uptake. Yet the anti-lipolytic action of insulin was markedly impaired in both cultured adipocytes and in mice injected with GSNO prior to administration of insulin. In cells, impaired ability of insulin to diminish phosphorylated PKA substrates in response to isoproterenol suggested impaired insulin-induced activation of PDE3B. Consistently, increased S-nitrosylation of PDE3B was detected in adipose tissue of high fat-fed obese mice. Site-directed mutagenesis revealed that Cys-768 and Cys-1040, two putative sites for S-nitrosylation adjacent to the substrate-binding site of PDE3B, accounted for ∼50% of its GSNO-induced S-nitrosylation. Collectively, PDE3B and the anti-lipolytic action of insulin may constitute novel targets for increased S-nitrosylation of adipose tissue in obesity.
Collapse
Affiliation(s)
- Hilla Ovadia
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Yulia Haim
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Ori Nov
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Orna Almog
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Julia Kovsan
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Nava Bashan
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096
| | - Assaf Rudich
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84103; National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84103, Israel.
| |
Collapse
|
39
|
Galli C, Passeri G, Macaluso GM. FoxOs, Wnts and oxidative stress-induced bone loss: new players in the periodontitis arena? J Periodontal Res 2011; 46:397-406. [PMID: 21332475 DOI: 10.1111/j.1600-0765.2011.01354.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Chronic periodontitis is a widespread disease affecting tooth-supporting structures that can lead to extensive loss of periodontal ligament and bone, ultimately resulting in tooth loss. Extensive evidence has demonstrated a strong association between age, metabolic disorders such as type II diabetes, oxidative stress and alveolar bone loss. The molecular players controlling bone maintenance and underlying age-related bone loss and its links to the general metabolism are currently the object of intense research. MATERIAL AND METHODS Recent findings are summarized to elucidate the molecular mechanisms linking oxidative stress, bone loss and metabolic factors. RESULTS It is well known that reactive oxygen species are an inevitable consequence of cellular respiration and that organisms have developed an efficient array of defenses against them. The core of this complex defense line is a family of transcription factors, known as FoxOs, which can bind to β-catenin and initiate a transcriptional programme regulating cell apoptosis, DNA repair and degradation of reactive oxygen species. An increase in reactive oxygen species due, for example, to age or insulin resistance, generates a situation in which bone formation is impaired by activation of FoxO, and a decrease in Wnt signaling and bone resorption are promoted. CONCLUSION The balance between FoxO and the Wnt pathway is finely tuned by systemic and local factors, creating a far-reaching mechanism that dictates the fate of mesenchymal progenitors and regulates the homeostasis of bone, providing a rationale for the impairment of systemic and alveolar bone maintenance clinically observed with age and metabolic diseases.
Collapse
Affiliation(s)
- C Galli
- Department of Internal Medicine Unit of Periodontology, University of Parma, Parma, Italy.
| | | | | |
Collapse
|
40
|
Shaik NA, Shaik JP, Ali S, Imran A, Rao DK. Increased frequency of micronuclei in diabetes mellitus patients using pioglitazone and glimepiride in combination. Food Chem Toxicol 2010; 48:3432-5. [PMID: 20868721 DOI: 10.1016/j.fct.2010.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Revised: 08/23/2010] [Accepted: 09/01/2010] [Indexed: 11/26/2022]
Abstract
Micronuclei frequency is a sensitive biomarker used to evaluate the genotoxicity induced by xenobiotics. Pioglitazone and glimepiride were associated with genotoxicity in experimental studies conducted in rats. Considering the lack of published reports on genotoxicity in T2DM patients using pioglitazone and glimepiride drugs in combination, current study aimed to assess whether the case and control groups significantly differ from each other in the frequency of micronuclei. Subjects comprise 127 T2DM patients (35-65 years old) under pioglitazone and glimepiride treatment for >5 years and control group of 140 age matched healthy controls (38-69 years old). Exfoliated oral mucosa cells were collected from buccal mucosa of all subjects and Feulgen/Fast-Green method was followed to screen for micronuclei. Factors such as gender, food habits, living areas and occupation have not shown significant association with the variation in micronuclei frequency among the studied subjects. However, T2DM patients under long term treatment of pioglitazone and glimepiride in combination, showed increased frequency of micronuclei as compared to controls (p<0.001). Current study suggests that the micronuclei assay can be used as a constituent among the panel of biomarkers to assess genotoxicity in T2DM patients under long term antihyperglycemic drug therapy.
Collapse
|
41
|
Berdichevsky A, Guarente L, Bose A. Acute oxidative stress can reverse insulin resistance by inactivation of cytoplasmic JNK. J Biol Chem 2010; 285:21581-9. [PMID: 20430894 DOI: 10.1074/jbc.m109.093633] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic oxidative stress results in decreased responsiveness to insulin, eventually leading to diabetes and cardiovascular disease. Activation of the JNK signaling pathway can mediate many of the effects of stress on insulin resistance through inhibitory phosphorylation of insulin receptor substrate 1. By contrast, exercise, which acutely increases oxidative stress in the muscle, improves insulin sensitivity and glucose tolerance in patients with Type 2 diabetes. To elucidate the mechanism underlying the contrasting effects of acute versus chronic oxidative stress on insulin sensitivity, we used a cellular model of insulin-resistant muscle to induce either chronic or acute oxidative stress and investigate their effects on insulin and JNK signaling. Chronic oxidative stress resulted in increased levels of phosphorylated (activated) JNK in the cytoplasm, whereas acute oxidative stress led to redistribution of JNK-specific phosphatase MKP7 from the nucleus into the cytoplasm, reduction in cytoplasmic phospho-JNK, and a concurrent accumulation of phospho-JNK in the nucleus. Acute oxidative stress restored normal insulin sensitivity and glucose uptake in insulin-resistant muscle cells, and this effect was dependent on MKP7. We propose that the contrasting effects of acute and chronic stress on insulin sensitivity are driven by changes in subcellular distribution of MKP7 and activated JNK.
Collapse
Affiliation(s)
- Alina Berdichevsky
- Novartis Institute for Biomedical Research, Cardiovascular and Metabolism Disease Area, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
42
|
Cooke MS, Evans MD, Mistry N, Lunec J. Role of dietary antioxidants in the prevention of in vivo oxidative DNA damage. Nutr Res Rev 2009; 15:19-42. [PMID: 19087397 DOI: 10.1079/nrr200132] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epidemiological evidence consistently shows that diets high in fresh fruit and vegetables significantly lower cancer risk. Given the postulated role of oxidative DNA damage in carcinogenesis, the assumption has been made that it is the antioxidant properties of food constituents, such as vitamin C, E and carotenoids, which confer protection. However, epidemiological studies with specific antioxidants, either singly or in combination, have not, on the whole, supported this hypothesis. In contrast, studies examining the in vitro effect of antioxidants upon oxidative DNA damage have generally been supportive, in terms of preventing damage induction. The same, however, cannot be said for the in vivo intervention studies where overall the results have been equivocal. Nevertheless, recent work has suggested that some dietary antioxidants may confer protective properties through a novel mechanism, unrelated to their conventional free-radical scavenging abilities. Upregulation of antioxidant defence, xenobiotic metabolism, or DNA-repair genes may all limit cellular damage and hence promote maintenance of cell integrity. However, until further work has clarified whether dietary supplementation with antioxidants confers a reduced risk of cancer and the mechanism by which this effect is exerted, the recommendation for a diet rich in fruit and vegetables remains valid empirically.
Collapse
Affiliation(s)
- M S Cooke
- Oxidative Stress Group, Division of Chemical Pathology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester LE2 7LX, UK.
| | | | | | | |
Collapse
|
43
|
Kisby GE, Kohama SG, Olivas A, Churchwell M, Doerge D, Spangler E, de Cabo R, Ingram DK, Imhof B, Bao G, Kow YW. Effect of caloric restriction on base-excision repair (BER) in the aging rat brain. Exp Gerontol 2009; 45:208-16. [PMID: 20005284 DOI: 10.1016/j.exger.2009.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Revised: 11/21/2009] [Accepted: 12/04/2009] [Indexed: 10/20/2022]
Abstract
Apyrimidinic/apurinic endonuclease (APE) is a key protein involved in the base-excision DNA repair (BER) pathway of oxidative DNA lesions. Using a novel oligonucleotide substrate, we demonstrate that APE activity in the frontal/parietal cortex (F/PCTX), cerebellum, brainstem, midbrain and hypothalamus declined with age in rats on an ad libitum (AL) diet. In contrast, APE activity for these brain regions was approximately 1.5-3 times higher in young, caloric restricted (CR) rats. Despite continuous CR treatment in all animals since six weeks of age, APE activity in the CR group started to decline by middle-age and continued into old age. However, CR maintained APE activity at a level that was significantly higher than that in AL rats across age and in the brain regions examined. Because Western analysis of APE, DNA polymerase beta and DNA ligase III levels in the F/PCTX of both CR and AL rats remained unchanged with age, this suggests that the increased APE activity in CR rats is the result of differential post-translational modification of APE.
Collapse
Affiliation(s)
- Glen E Kisby
- Center for Research on Occupational and Environmental Toxicology, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Telomerase is a specialized reverse transcriptase that is responsible for extending and preserving the end of the chromosomes (telomeres). Telomerase plays a key role in regulating the lifespan of mammalian cells and is involved in critical aspects of cellular ageing processes. In this review, we will briefly summarize our current understanding of the functions of telomeres, telomerase and their regulation. Considering that compensatory islet hyperplasia and beta-cell regeneration play important roles in the prevention and/or delay of the onset of overt diabetes, we will also examine current literature regarding the effects of diabetes on telomere shortening and provide insights from our own studies on the role of telomerase in beta-cell regeneration.
Collapse
Affiliation(s)
- C W Liew
- Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
45
|
Prooxidative effects of green tea polyphenol (−)-epigallocatethin-3-gallate on the HIT-T15 pancreatic beta cell line. Cell Biol Toxicol 2009; 26:189-99. [DOI: 10.1007/s10565-009-9137-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 08/28/2009] [Indexed: 11/26/2022]
|
46
|
Singh RB, Khanna HK, Niaz MA. Randomized, Double-blind Placebo-controlled Trial of Coenzyme Q10 in Chronic Renal Failure: Discovery of a New Role. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/13590840020013266] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
47
|
Bashan N, Kovsan J, Kachko I, Ovadia H, Rudich A. Positive and negative regulation of insulin signaling by reactive oxygen and nitrogen species. Physiol Rev 2009; 89:27-71. [PMID: 19126754 DOI: 10.1152/physrev.00014.2008] [Citation(s) in RCA: 370] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Regulated production of reactive oxygen species (ROS)/reactive nitrogen species (RNS) adequately balanced by antioxidant systems is a prerequisite for the participation of these active substances in physiological processes, including insulin action. Yet, increasing evidence implicates ROS and RNS as negative regulators of insulin signaling, rendering them putative mediators in the development of insulin resistance, a common endocrine abnormality that accompanies obesity and is a risk factor of type 2 diabetes. This review deals with this dual, seemingly contradictory, function of ROS and RNS in regulating insulin action: the major processes for ROS and RNS generation and detoxification are presented, and a critical review of the evidence that they participate in the positive and negative regulation of insulin action is provided. The cellular and molecular mechanisms by which ROS and RNS are thought to participate in normal insulin action and in the induction of insulin resistance are then described. Finally, we explore the potential usefulness and the challenges in modulating the oxidant-antioxidant balance as a potentially promising, but currently disappointing, means of improving insulin action in insulin resistance-associated conditions, leading causes of human morbidity and mortality of our era.
Collapse
Affiliation(s)
- Nava Bashan
- Department of Clinical Biochemistry, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | |
Collapse
|
48
|
Amenta F, Traini E, Tomassoni D, Mignini F. Pharmacokinetics of different formulations of tioctic (alpha-lipoic) acid in healthy volunteers. Clin Exp Hypertens 2009; 30:767-75. [PMID: 19021026 DOI: 10.1080/10641960802563568] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This study was designed to evaluate in healthy volunteers plasma and cellular (in erythrocytes) of three formulations of alpha-lipoic acid (ALA) available in Italy with different rates of absorption, two with a claimed high absorption rate (Byodinoral 600 QR, Tiocronal 600 HR) and one with a claimed prolonged absorption rate (Tiobec 600 retard). These formulations were compared with the registered ethic formulation of the compound (Thioctacid 600 mg HR), available in Germany. Area under the curve from time 0 to last measured time (AUC(t)), peak plasma concentration (C(max)) of ALA, and time (T(max) ) at which C(max) was observed were the plasma kinetic parameters measured. Concentration of ALA at different sampling times was the only parameter assessed for erythrocytes. The AUC(t) values were similar for the four formulations of ALA tested. C(max) was significantly higher for Byodinoral 600 QR, Tiocronal 600 HR compared to Thioctacid 600 mg HR or Tiobec 600 retard, whereas T(max) value was significantly shorter for Byodinoral 600 QR in the order by Tiocronal 600 HR, Thioctacid 600 mg HR, and Tiobec 600 retard. ALA concentrations that accumulated in erytrocytes after the administration of the different formulations of the antioxidant are directly proportional to the plasma levels of each formulation. Because antioxidant capabilities of ALA depend on the glutathione regeneration the compound induces in cells, the most rationale approach for eliciting antioxidant activity at the cellular level is probably in the use of a formulation allowing the compound to reach its target at highest concentrations and in the shortest time.
Collapse
Affiliation(s)
- Francesco Amenta
- Centro di Ricerche Cliniche, Dipartimento di Medicina Sperimentale e Sanità Pubblica, Università di Camerino, Camerino, Italy.
| | | | | | | |
Collapse
|
49
|
Iwahashi H. 8-Oxo-7,8-dihydro-2'-deoxyguanosine Forms a Relatively Unstable Tetrameric Structure Compared with 2'-Deoxyguanosine. J Clin Biochem Nutr 2009; 44:57-61. [PMID: 19177189 PMCID: PMC2613500 DOI: 10.3164/jcbn.08-157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Accepted: 07/25/2008] [Indexed: 11/22/2022] Open
Abstract
The hydrogen-bonded guanine tetrad, or G-quartet has been implicated in a variety of biological roles, including the function of chromosome telomeres. Here effect of the hydroxylation of guanosine at the 8 position on the G-quartet formation was examined. Electrospray inonization mass (ESI-MS) spectra of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) and 2'-deoxyguanosine (dG) were measured in order to know whether or not 8-oxodG forms a tetrameric structure as 2'-deoxyguanosine forms in teromeres. The ESI-MS spectra of dG shows prominent peaks at m/z 290, m/z 557, and m/z 1092, corresponding to [dG + Na]+, [dG2 + Na]+, and [dG4 + Na]+ in the presence of 0.1 mM NaCl. On the other hand, the ESI-MS spectra of 8-oxodG in the presence of 0.1 mM NaCl shows prominent peaks at m/z 306 and m/z 589, corresponding to [8-oxodG + Na]+ and [8-oxodG2 + Na]+. The results showed that 8-oxodG forms a relatively unstable tetrameric structure compared with dG.
Collapse
Affiliation(s)
- Hideo Iwahashi
- *To whom correspondence should be addressed. Tel: +81-73-441-0772 Fax: +81-73-441-0772 E-mail:
| |
Collapse
|
50
|
Kuroda E, Horikawa Y, Enya M, Oda N, Suzuki E, Iizuka K, Takeda J. Identification of minimal promoter and genetic variants of Kruppel-like factor 11 gene and association analysis with type 2 diabetes in Japanese. Endocr J 2009; 56:275-86. [PMID: 19122346 DOI: 10.1507/endocrj.k08e-302] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Genetic analysis of the KLF11 gene revealed two rare variants, A347S and T220M, segregating in families with early-onset type 2 diabetes, and one frequent polymorphic Q62R variant significantly associated with type 2 diabetes in Northern Europeans. Furthermore, it has been reported that over-expression of KLF11 has a deleterious effect on insulin promoter activity. Thus, an altered expression level of KLF11 may contribute to the occurrence of type 2 diabetes. To investigate the contribution of KLF11 to type 2 diabetes in Japanese, we surveyed the 5' flanking region of KLF11 by reporter assay and identified the minimal promoter region of the gene. The promoter region from -250 to +162 bp including five Sp1 binding sites showed basal promoter activity both in MIN6-m9 and HepG2 cells. We also examined the entire region of KLF11 to detect genetic variants. A total of 19 polymorphisms, six of which are novel, were identified, but none of them showed association with the occurrence of type 2 diabetes. Two of the identified polymorphisms, R29Q and S124F, are novel coding variants. Functional analyses of these variants were performed, and similarly reduced effects on transcriptional activities of insulin, catalase1, and the Smad7 gene were found. We conclude that variants of KLF11 are not a major factor in the occurrence of type 2 diabetes in Japanese. The promoter region of KLF11 identified in the present study should be useful in further elucidation of the transcriptional regulation mechanism of the gene and genetic analyses of type 2 diabetes.
Collapse
Affiliation(s)
- Eiji Kuroda
- Department of Diabetes and Endocrinology, Division of Molecule and Structure, Gifu University School of Medicine, Gifu, Japan
| | | | | | | | | | | | | |
Collapse
|