1
|
Tshivhase AM, Matsha T, Raghubeer S. The protective role of resveratrol against high glucose-induced oxidative stress and apoptosis in HepG2 cells. Food Sci Nutr 2024; 12:3574-3584. [PMID: 38726423 PMCID: PMC11077230 DOI: 10.1002/fsn3.4027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/19/2023] [Accepted: 01/31/2024] [Indexed: 05/12/2024] Open
Abstract
High glucose concentrations result in oxidative stress, leading to damage of cellular constituents like DNA, proteins, and lipids, ultimately resulting in apoptosis. Resveratrol, a polyphenol phytoalexin, has been studied for its potential therapeutic effects on diabetes. This study investigated the influence of high glucose (HG) on HepG2 cells and assessed resveratrol's effect on high-glucose-induced oxidative stress and apoptosis. HepG2 cells were cultured for 48 and 72 h with high glucose (40 mM), low resveratrol (25 μM), high resveratrol (50 μM), high glucose + low resveratrol, and high glucose + high resveratrol. After exposure, oxidative and apoptosis-related gene expression was evaluated using quantitative polymerase chain reaction (qPCR), and lactate dehydrogenase (LDH) release was measured using the supernatant. In HepG2 cells cultured with high glucose, all antioxidant enzymes (SOD, superoxide dismutase; GPx1, glutathione peroxidase 1; CAT, catalase; Nrf2, nuclear factor erythroid 2-related factor 2; and NQO1, NAD(P)H quinone oxidoreductase 1) were significantly reduced; however, when HepG2 cells were cultured with resveratrol (25 and 50 μM) and high glucose, the expression levels of all antioxidant enzymes were increased. The anti-apoptotic gene (B-cell lymphoma 2; Bcl2) and the DNA repair gene (Oxoguanine glycosylase-1, OGG1) were significantly decreased following high glucose exposure to HepG2 cells. Surprisingly, the expression levels of Bcl2 and OGG1 were notably elevated after resveratrol treatment. Furthermore, high glucose levels increased the LHD release in HepG2 cells, whereas resveratrol treatment reduced the LDH release. Our results demonstrate that resveratrol provides protection against oxidative stress and apoptosis induced by high glucose in HepG2 cells. Hence, resveratrol shows potential as an effective approach to address the impaired antioxidant response resulting from elevated glucose levels commonly observed in diabetes and metabolic disorders.
Collapse
Affiliation(s)
- Abegail Mukhethwa Tshivhase
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness SciencesCape Peninsula University of TechnologyBellvilleSouth Africa
| | - Tandi Matsha
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness SciencesCape Peninsula University of TechnologyBellvilleSouth Africa
- Sefako Makgatho Health Sciences UniversityGa‐RankuwaSouth Africa
| | - Shanel Raghubeer
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness SciencesCape Peninsula University of TechnologyBellvilleSouth Africa
| |
Collapse
|
2
|
Hassan WM. Oxidative DNA Damage and Zinc Status in Patients With Rheumatoid Arthritis in Duhok, Iraq. Cureus 2024; 16:e52860. [PMID: 38406004 PMCID: PMC10886431 DOI: 10.7759/cureus.52860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/27/2024] Open
Abstract
Background In patients with rheumatoid arthritis, oxidative DNA damage is increased by deficient zinc levels as well as increasing disease activity. However, the relationship between zinc levels, disease activity, and oxidative DNA damage remains unclear. In this study, we investigated serum zinc levels and disease activity and their association with 8-hydroxy-2-deoxyguanosine (8-OHdG). Methodology This case-control study was conducted among rheumatoid arthritis patients (n = 264) and healthy individuals (n = 192). Oxidative DNA damage was assessed by measuring serum 8-OHdG using enzyme-linked immunosorbent assay. Colorimetry was used to measure serum zinc levels. Disease activity was assessed using the Disease Activity Score-28 (DAS-28) score. Results Significantly higher 8-OHdG levels (p < 0.00) were found in the test group compared to the control group. Moreover, significantly lower serum zinc levels (p < 0.001) were noted in patients with rheumatoid arthritis compared to the control group. In addition, higher 8-OHdG levels were found in patients with low serum zinc levels compared to those with normal mean serum zinc levels. Lower levels of DNA oxidative damage were found in patients with moderate and low disease activity compared to those with high disease activity. A significant negative correlation was noted between serum zinc levels and DAS-28 scores and oxidative DNA damage marker (r = - 0.30, p = 0.038 and r = - 0.26, p = 0.043, respectively), while a significant positive correlation was observed between body mass index and 8-OHdG (r = 0.22, p = 0.02) in healthy individuals. Conclusions High serum 8-OHdG levels and high disease activity with low mean serum zinc levels may indicate a high degree of oxidative DNA damage in patients with rheumatoid arthritis.
Collapse
|
3
|
Krawczyk M, Burzynska-Pedziwiatr I, Wozniak LA, Bukowiecka-Matusiak M. Impact of Polyphenols on Inflammatory and Oxidative Stress Factors in Diabetes Mellitus: Nutritional Antioxidants and Their Application in Improving Antidiabetic Therapy. Biomolecules 2023; 13:1402. [PMID: 37759802 PMCID: PMC10526737 DOI: 10.3390/biom13091402] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by hyperglycaemia and oxidative stress. Oxidative stress plays a crucial role in the development and progression of diabetes and its complications. Nutritional antioxidants derived from dietary sources have gained significant attention due to their potential to improve antidiabetic therapy. This review will delve into the world of polyphenols, investigating their origins in plants, metabolism in the human body, and relevance to the antioxidant mechanism in the context of improving antidiabetic therapy by attenuating oxidative stress, improving insulin sensitivity, and preserving β-cell function. The potential mechanisms of, clinical evidence for, and future perspectives on nutritional antioxidants as adjuvant therapy in diabetes management are discussed.
Collapse
|
4
|
Huang D, Shin WK, De la Torre K, Lee HW, Min S, Shin A, Lee JK, Kang D. Association between metabolic syndrome and gastric cancer risk: results from the Health Examinees Study. Gastric Cancer 2023; 26:481-492. [PMID: 37010633 DOI: 10.1007/s10120-023-01382-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/03/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Previous studies suggested that metabolic syndrome (MetS) might create a pro-cancer environment and increase cancer incidence. However, evidence on the risk of gastric cancer (GC) was limited. This study aimed to evaluate the association between MetS and its components and GC in the Korean population. METHODS Included were 108,397 individuals who participated in the large-scale prospective cohort study, the Health Examinees-Gem study during 2004-2017. The multivariable Cox proportional was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) on the association between MetS and its components with GC risk. Age was used as the time scale in the analyses. The stratified analysis was performed to determine the joint effect of lifestyle factors and MetS on GC risk in different groups. RESULTS During the mean follow-up of 9.1 years, 759 cases of newly diagnosed cancer (408 men and 351 women) were identified. Overall, participants with MetS had a 26% increased risk of GC than those without MetS (HR 1.26; 95% CI 1.07-1.47); the risk increased with the number of MetS components (p for trend 0.01). Hypertriglyceridemia, low HDL-cholesterol, and hyperglycemia were independently associated with the risk of GC. The potential joint effect of MetS and current smokers (p for interaction 0.02) and obesity (BMI ≥ 25.0) (p for interaction 0.03) in GC. CONCLUSIONS In this prospective cohort study, we found that MetS were associated with an increased risk of GC in the Korean population. Our findings suggest that MetS may be a potentially modifiable risk factor for GC risk.
Collapse
Affiliation(s)
- Dan Huang
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Woo-Kyoung Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Katherine De la Torre
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Hwi-Won Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Sukhong Min
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
- Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Jong-Koo Lee
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Daehee Kang
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea.
| |
Collapse
|
5
|
Rizwan H, Satapathy SS, Si S, Kumar S, Kumari G, Pal A. Effect of Au@SiO 2 core shell nanoparticles on HG-induced oxidative stress triggered apoptosis in keratinocytes. Life Sci 2023; 328:121893. [PMID: 37392778 DOI: 10.1016/j.lfs.2023.121893] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/17/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Growing evidences suggest that excess generation of highly reactive free oxygen/nitrogen radicals (ROS/RNS) are largely due to hyperglycemia causes oxidative stress. Further, excess accumulation of ROS/RNS in cellular compartments aggravates the development and progression of diabetes and its associated complications. Impaired wound healing in diabetic condition is a known vital complication all around the world. Thus, an antioxidant agent having the potential for hindering the oxidative/nitrosative stress triggered diabetic skin complication is required. The present investigation was carried out to understand the impact of silica coated gold nanoparticle (Au@SiO2 NPs) on high glucose (HG)-induced keratinocyte complications. We demonstrated that HG environment enhanced the ROS and RNS accumulations and reduced in cellular antioxidant capacities in keratinocte cells, however, Au@SiO2 NPs treatment restored the HG effect. Furthermore, excess production of ROS/RNS was associated with mitochondrial dysfunction, characterized by loss of mitochondrial membrane potential (ΔΨm), and increased in mitochondrial mass, which was restored by Au@SiO2 NPs treatment in keratinocyte cells. In addition, HG-induced excess production of ROS/RNA caused an increased in the biomolecules damage including lipid peroxidation (LPO), and protein carbonylation (PC), 8-oxoguanine DNA glycosylase-1 (OGG1) expression and increased 8-hydroxydeoxyguanosine (8-OHdG) accumulations in DNA, leading to activation of ERK1/2MAPK, AKT and tuberin pathway, inflammatory reaction, and finally apoptotic cell death. In conclusion, our findings showed that Au@SiO2 NPs treatment improved the HG-induced keratinocytes injury by suppressing the oxidative/nitrosative stress, elevating the antioxidant defence system, thereby inhibiting the inflammatory mediators and apoptosis, which may be a therapeutic cure for the diabetic keratinocyte problems.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Smith Sagar Satapathy
- School of Chemical Technology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Satyabrata Si
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; School of Chemical Technology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Sonu Kumar
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Golden Kumari
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Arttatrana Pal
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India.
| |
Collapse
|
6
|
Zhao X, Ma Y, Shi M, Huang M, Xin J, Ci S, Chen M, Jiang T, Hu Z, He L, Pan F, Guo Z. Excessive iron inhibits insulin secretion via perturbing transcriptional regulation of SYT7 by OGG1. Cell Mol Life Sci 2023; 80:159. [PMID: 37209177 PMCID: PMC11072990 DOI: 10.1007/s00018-023-04802-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/22/2023]
Abstract
Although iron overload is closely related to the occurrence of type 2 diabetes mellitus (T2DM), the specific mechanism is unclear. Here, we found that excessive iron inhibited the secretion of insulin (INS) and impaired islet β cell function through downregulating Synaptotagmin 7 (SYT7) in iron overload model in vivo and in vitro. Our results further demonstrated that 8-oxoguanine DNA glycosylase (OGG1), a key protein in the DNA base excision repair, was an upstream regulator of SYT7. Interestingly, such regulation could be suppressed by excessive iron. Ogg1-null mice, iron overload mice and db/db mice exhibit reduced INS secretion, weakened β cell function and subsequently impaired glucose tolerance. Notably, SYT7 overexpression could rescue these phenotypes. Our data revealed an intrinsic mechanism by which excessive iron inhibits INS secretion through perturbing the transcriptional regulation of SYT7 by OGG1, which suggested that SYT7 was a potential target in clinical therapy for T2DM.
Collapse
Affiliation(s)
- Xingqi Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Ying Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Munan Shi
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Miaoling Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Jingyu Xin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Shusheng Ci
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Meimei Chen
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China
| | - Tao Jiang
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China.
| |
Collapse
|
7
|
Nikfarjam S, Singh KK. DNA damage response signaling: A common link between cancer and cardiovascular diseases. Cancer Med 2023; 12:4380-4404. [PMID: 36156462 PMCID: PMC9972122 DOI: 10.1002/cam4.5274] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/10/2022] [Accepted: 07/19/2022] [Indexed: 11/10/2022] Open
Abstract
DNA damage response (DDR) signaling ensures genomic and proteomic homeostasis to maintain a healthy genome. Dysregulation either in the form of down- or upregulation in the DDR pathways correlates with various pathophysiological states, including cancer and cardiovascular diseases (CVDs). Impaired DDR is studied as a signature mechanism for cancer; however, it also plays a role in ischemia-reperfusion injury (IRI), inflammation, cardiovascular function, and aging, demonstrating a complex and intriguing relationship between cancer and pathophysiology of CVDs. Accordingly, there are increasing number of reports indicating higher incidences of CVDs in cancer patients. In the present review, we thoroughly discuss (1) different DDR pathways, (2) the functional cross talk among different DDR mechanisms, (3) the role of DDR in cancer, (4) the commonalities and differences of DDR between cancer and CVDs, (5) the role of DDR in pathophysiology of CVDs, (6) interventional strategies for targeting genomic instability in CVDs, and (7) future perspective.
Collapse
Affiliation(s)
- Sepideh Nikfarjam
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Krishna K Singh
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
8
|
Nuclear factor Nrf2 promotes glycosidase OGG1 expression by activating the AKT pathway to enhance leukemia cell resistance to cytarabine. J Biol Chem 2022; 299:102798. [PMID: 36528059 PMCID: PMC9823221 DOI: 10.1016/j.jbc.2022.102798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Chemotherapy resistance is the dominant challenge in the treatment of acute myeloid leukemia (AML). Nuclear factor E2-related factor 2 (Nrf2) exerts a vital function in drug resistance of many tumors. Nevertheless, the potential molecular mechanism of Nrf2 regulating the base excision repair pathway that mediates AML chemotherapy resistance remains unclear. Here, in clinical samples, we found that the high expression of Nrf2 and base excision repair pathway gene encoding 8-hydroxyguanine DNA glycosidase (OGG1) was associated with AML disease progression. In vitro, Nrf2 and OGG1 were highly expressed in drug-resistant leukemia cells. Upregulation of Nrf2 in leukemia cells by lentivirus transfection could decrease the sensitivity of leukemia cells to cytarabine, whereas downregulation of Nrf2 in drug-resistant cells could enhance leukemia cell chemosensitivity. Meanwhile, we found that Nrf2 could positively regulate OGG1 expression in leukemia cells. Our chromatin immunoprecipitation assay revealed that Nrf2 could bind to the promoter of OGG1. Furthermore, the use of OGG1 inhibitor TH5487 could partially reverse the inhibitory effect of upregulated Nrf2 on leukemia cell apoptosis. In vivo, downregulation of Nrf2 could increase the sensitivity of leukemia cell to cytarabine and decrease OGG1 expression. Mechanistically, Nrf2-OGG1 axis-mediated AML resistance might be achieved by activating the AKT signaling pathway to regulate downstream apoptotic proteins. Thus, this study reveals a novel mechanism of Nrf2-promoting drug resistance in leukemia, which may provide a potential therapeutic target for the treatment of drug-resistant/refractory leukemia.
Collapse
|
9
|
Hong X, Hu Y, Yuan Z, Fang Z, Zhang X, Yuan Y, Guo C. Oxidatively Damaged Nucleic Acid: Linking Diabetes and Cancer. Antioxid Redox Signal 2022; 37:1153-1167. [PMID: 35946074 DOI: 10.1089/ars.2022.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Our current knowledge of the mechanism between diabetes and cancer is limited. Oxidatively damaged nucleic acid is considered a critical factor to explore the connections between these two diseases. Recent Advances: The link between diabetes mellitus and cancer has attracted increasing attention in recent years. Emerging evidence supports that oxidatively damaged nucleic acid caused by an imbalance between reactive oxygen species generation and elimination is a bridge connecting diabetes and cancer. 8-Oxo-7,8-dihydro-2'-deoxyguanosine and 8-oxo-7,8-dihydroguanosine assume important roles as biomarkers in assessing the relationship between oxidatively damaged nucleic acid and cancer. Critical Issues: The consequences of diabetes are extensive and may lead to the occurrence of cancer by influencing a combination of factors. At present, there is no direct evidence that diabetes causes cancer by affecting a single factor. Furthermore, the difficulty in controlling variables and differences in detection methods lead to poor reliability and repeatability of results, and there are no clear cutoff values for biomarkers to indicate cancer risk. Future Directions: A better understanding of connections as well as mechanisms between diabetes and cancer is still needed. Both diabetes and cancer are currently intractable diseases. Further exploration of the specific mechanism of oxidatively damaged nucleic acid in the connection between diabetes and cancer is urgently needed. In the future, it is necessary to further take oxidatively damaged nucleic acid as an entry point to provide new ideas for the diagnosis and treatment of diabetes and cancer. Experimental drugs targeting the repair process of oxidatively generated damage require an extensive preclinical evaluation and could ultimately provide new treatment strategies for these diseases. Antioxid. Redox Signal. 37, 1153-1167.
Collapse
Affiliation(s)
- Xiujuan Hong
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqiu Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijun Yuan
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihao Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Cheng Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Rizwan H, Kumar S, Kumari G, Pal A. High glucose-induced increasing reactive nitrogen species accumulation triggered mitochondrial dysfunction, inflammation, and apoptosis in keratinocytes. Life Sci 2022; 312:121208. [PMID: 36427546 DOI: 10.1016/j.lfs.2022.121208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022]
Abstract
Growing evidence indicates that skin injuries are a common complication of diabetes. However, the cellular and molecular mechanisms of high glucose (HG) environment trigger nitrosative stress-mediated inflammation and apoptosis in keratinocytes remains unknown. Here we investigated whether reactive nitrogen species (RNS) induced by HG environment restrain antioxidant activity, and mitochondrial dysfunction leading to inflammation, and apoptosis via stress signaling pathways in keratinocytes. Our results established that the HG environment enhanced the production of nitric oxide (NO) and peroxynitrite anion (ONOO-) by inducible NO synthase (iNOS) in keratinocytes. Overproduction of RNS in HG environment suppress the antioxidants activity leading to mitochondrial dysfunction, characterized by loss of mitochondrial membrane potential (ΔΨm), increase in mitochondrial mass, decrease in mitochondrial transcription factor A(TFAM), increase in mitochondrial DNA (mtDNA) displacement loop (D-loop) and decrease in glycolytic flux concentration, which was attenuated by pharmacological inhibitors of NO/ONOO-, Nω-Nitro-l-argininemethyl ester hydrochloride (NAME)/hydralazine hydrochloride (Hyd.HCl). Excess production of RNS in HG environment restrained 8-oxoguanine DNA glycosylase-1 (OGG1) expression and increased 8-hydroxydeoxyguanosine (8-OHdG) accumulations in DNA were regulated by NO or ONOO-. Further, HG-induced RNA production caused an increase in the production of inflammatory mediators accompanied by activation of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3 signaling cascade, lipid peroxidation (LPO), and protein carbonylation (PC) reactions followed by breakdown the cell-cell communication and apoptosis. Pre-treatment of cell with NAME/Hyd.HCl, diminished the expression of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3, inflammatory mediators, and attenuated apoptosis in keratinocytes. Together, our results indicated that excess production of RNS in HG environment triggered inflammation and apoptosis, mediated by activation of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3 signaling cascades in keratinocytes.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Sonu Kumar
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Golden Kumari
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Arttatrana Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India.
| |
Collapse
|
11
|
Inhibition of 8-oxoguanine DNA glycosylase (OGG1) expression suppresses polycystic ovarian syndrome via the NF-κB signaling pathway. Reprod Biol 2022; 22:100679. [PMID: 35961097 DOI: 10.1016/j.repbio.2022.100679] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/06/2022] [Accepted: 07/25/2022] [Indexed: 11/20/2022]
Abstract
It has been reported that oxidative stress and chronic inflammation may be involved in the pathogenesis of polycystic ovary syndrome (PCOS). 8-oxoguanine DNA glycosylase (OGG1) is the main glycosylase that catalyzes the excision of DNA oxidation products. In this study, we investigated the role and potential mechanisms of OGG1 in the development of PCOS. We first analyzed OGG1 levels in serum and follicular fluid (FF) of PCOS patients, and significantly elevated OGG1 levels were noted in PCOS patients. We similarly observed a significant upregulation of OGG1 expression levels in ovarian tissue of the dehydroepiandrosterone (DHEA)-induced PCOS rat model. In addition, increased apoptosis and increased production of reactive oxygen species (ROS) were observed after the addition of OGG1-specific inhibitor (TH5487) in human granulosa-like tumor cell line (KGN) cells following a concentration gradient, along with a significant decrease in mRNA levels of inflammatory factors such as CXCL2, IL-6, MCP1, IL-1β, and IL-18. Significant decreases in protein phosphorylation levels of P65 and IκBα were also observed in cells. In addition, we found a significant positive correlation between OGG1 and IL-6 expression levels in human and DHEA-induced PCOS rat models. In conclusion, our results suggest that OGG1 might be involved in the pathogenesis of PCOS by regulating the secretion of IL-6 through NF-κB signaling pathway, and there might be a balance between the inhibition of oxidative stress and the promotion of chronic inflammation by OGG1 on KGN cells.
Collapse
|
12
|
Small molecule-mediated allosteric activation of the base excision repair enzyme 8-oxoguanine DNA glycosylase and its impact on mitochondrial function. Sci Rep 2022; 12:14685. [PMID: 36038587 PMCID: PMC9424235 DOI: 10.1038/s41598-022-18878-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/22/2022] [Indexed: 02/07/2023] Open
Abstract
8-Oxoguanine DNA glycosylase (OGG1) initiates base excision repair of the oxidative DNA damage product 8-oxoguanine. OGG1 is bifunctional; catalyzing glycosyl bond cleavage, followed by phosphodiester backbone incision via a β-elimination apurinic lyase reaction. The product from the glycosylase reaction, 8-oxoguanine, and its analogues, 8-bromoguanine and 8-aminoguanine, trigger the rate-limiting AP lyase reaction. The precise activation mechanism remains unclear. The product-assisted catalysis hypothesis suggests that 8-oxoguanine and analogues bind at the product recognition (PR) pocket to enhance strand cleavage as catalytic bases. Alternatively, they may allosterically activate OGG1 by binding outside of the PR pocket to induce an active-site conformational change to accelerate apurinic lyase. Herein, steady-state kinetic analyses demonstrated random binding of substrate and activator. 9-Deazaguanine, which can't function as a substrate-competent base, activated OGG1, albeit with a lower Emax value than 8-bromoguanine and 8-aminoguanine. Random compound screening identified small molecules with Emax values similar to 8-bromoguanine. Paraquat-induced mitochondrial dysfunction was attenuated by several small molecule OGG1 activators; benefits included enhanced mitochondrial membrane and DNA integrity, less cytochrome c translocation, ATP preservation, and mitochondrial membrane dynamics. Our results support an allosteric mechanism of OGG1 and not product-assisted catalysis. OGG1 small molecule activators may improve mitochondrial function in oxidative stress-related diseases.
Collapse
|
13
|
Zhang L, Zhang Y, Qin X, Jiang X, Zhang J, Mao L, Jiang Z, Jiang Y, Liu G, Qiu J, Chen C, Qiu F, Zou Z. Recombinant ACE2 protein protects against acute lung injury induced by SARS-CoV-2 spike RBD protein. Crit Care 2022; 26:171. [PMID: 35681221 PMCID: PMC9178547 DOI: 10.1186/s13054-022-04034-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/27/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infection leads to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Both clinical data and animal experiments suggest that the renin-angiotensin system (RAS) is involved in the pathogenesis of SARS-CoV-2-induced ALI. Angiotensin-converting enzyme 2 (ACE2) is the functional receptor for SARS-CoV-2 and a crucial negative regulator of RAS. Recombinant ACE2 protein (rACE2) has been demonstrated to play protective role against SARS-CoV and avian influenza-induced ALI, and more relevant, rACE2 inhibits SARS-CoV-2 proliferation in vitro. However, whether rACE2 protects against SARS-CoV-2-induced ALI in animal models and the underlying mechanisms have yet to be elucidated. METHODS AND RESULTS Here, we demonstrated that the SARS-CoV-2 spike receptor-binding domain (RBD) protein aggravated lipopolysaccharide (LPS)-induced ALI in mice. SARS-CoV-2 spike RBD protein directly binds and downregulated ACE2, leading to an elevation in angiotensin (Ang) II. AngII further increased the NOX1/2 through AT1R, subsequently causing oxidative stress and uncontrolled inflammation and eventually resulting in ALI/ARDS. Importantly, rACE2 remarkably reversed SARS-CoV-2 spike RBD protein-induced ALI by directly binding SARS-CoV-2 spike RBD protein, cleaving AngI or cleaving AngII. CONCLUSION This study is the first to prove that rACE2 plays a protective role against SARS-CoV-2 spike RBD protein-aggravated LPS-induced ALI in an animal model and illustrate the mechanism by which the ACE2-AngII-AT1R-NOX1/2 axis might contribute to SARS-CoV-2-induced ALI.
Collapse
Affiliation(s)
- Lingbing Zhang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yandan Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Ziqi Jiang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yu Jiang
- Department of Respiratory Medicine, The University-Town Hospital of Chongqing Medical University, Chongqing, 401331, People's Republic of China
| | - Gang Liu
- Department of Emergency, The University-Town Hospital of Chongqing Medical University, Chongqing, 401331, People's Republic of China
| | - Jingfu Qiu
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Chengzhi Chen
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
14
|
Liu Q, Cui Y, Ding N, Zhou C. Knockdown of circ_0003928 ameliorates high glucose-induced dysfunction of human tubular epithelial cells through the miR-506-3p/HDAC4 pathway in diabetic nephropathy. Eur J Med Res 2022; 27:55. [PMID: 35392987 PMCID: PMC8991937 DOI: 10.1186/s40001-022-00679-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/20/2022] [Indexed: 11/10/2022] Open
Abstract
Background Previous data have indicated the importance of circular RNA (circRNA) in the pathogenesis of diabetic nephropathy (DN). The study is designed to investigate the effects of circ_0003928 on oxidative stress and apoptosis of high glucose (HG)-treated human tubular epithelial cells (HK-2) and the underlying mechanism. Methods The DN cell model was established by inducing HK-2 cells using 30 mmol/L D-glucose. RNA expression of circ_0003928, miR-506-3p and histone deacetylase 4 (HDAC4) was detected by quantitative real-time polymerase chain reaction. Cell viability and proliferation were investigated by cell counting kit-8 and 5-Ethynyl-29-deoxyuridine (EdU) assays, respectively. Oxidative stress was evaluated by commercial kits. Caspase 3 activity and cell apoptotic rate were assessed by a caspase 3 activity assay and flow cytometry analysis, respectively. Protein expression was detected by Western blotting analysis. The interactions among circ_0003928, miR-506-3p and HDAC4 were identified by dual-luciferase reporter and RNA pull-down assays. Results Circ_0003928 and HDAC4 expression were significantly upregulated, while miR-506-3p was downregulated in the serum of DN patients and HG-induced HK-2 cells. HG treatment inhibited HK-2 cell proliferation, but induced oxidative stress and cell apoptosis; however, these effects were reversed after circ_0003928 depletion. Circ_0003928 acted as a miR-506-3p sponge, and HDAC4 was identified as a target gene of miR-506-3p. Moreover, the circ_0003928/miR-506-3p/HDAC4 axis regulated HG-induced HK-2 cell dysfunction. Conclusion Circ_0003928 acted as a sponge for miR-506-3p to regulate HG-induced oxidative stress and apoptosis of HK-2 cells through HDAC4, which suggested that circ_0003928 might be helpful in the therapy of DN. Supplementary Information The online version contains supplementary material available at 10.1186/s40001-022-00679-y.
Collapse
Affiliation(s)
- Qiong Liu
- Department of Nephrology, Hebei General Hospital, Shijiazhuang, China
| | - Yuanyuan Cui
- Department of Endocrine Rheumatology and Immunology, People's Hospital of Gaotang County, Gaotang, China
| | - Nan Ding
- Department of Clinical Laboratory, Hebei General Hospital, Shijiazhuang, China
| | - Changxue Zhou
- Department of Kidney Internal Medicine, Zaozhuang Municipal Hospital, No. 41 Longtou Road, Central District, Zaozhuang, 277100, China.
| |
Collapse
|
15
|
Kanbay M, Copur S, Demiray A, Tuttler KR. Cardiorenal Metabolic Consequences of Nighttime Snacking: Is it an Innocent Eating Behavior? Curr Nutr Rep 2022; 11:347-353. [PMID: 35195849 DOI: 10.1007/s13668-022-00403-6] [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] [Accepted: 02/02/2022] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Health consequences of nighttime eating, as a publicly discussed eating behavior type, have been speculated lately. Nighttime eating has been linked to various metabolic outcomes including hyperlipidemia, hypertriglyceridemia, hyperglycemia, weight gain, elevated blood pressure, obesity, and metabolic syndrome, and cardiorenal outcomes such as atherosclerosis, a decline in eGFR, and proteinuria. RECENT FINDINGS Although the exact underlying pathophysiological mechanism is not yet clear, multiple hypotheses including disrupted circadian rhythm, altered hormonal levels, and decline in cellular regeneration have been proposed. In this review, we aim to evaluate the growing literature on nighttime eating behavior in terms of metabolic and cardiorenal outcomes, pathophysiological basis, and potential therapeutic alternatives.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, 34010, Istanbul, Turkey.
| | - Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Kathherine R Tuttler
- Division of Nephrology, University of Washington, Seattle, WA, USA
- Providence Medical Research Center, Providence Health Care, Spokane, WA, USA
| |
Collapse
|
16
|
Lima JEBF, Moreira NCS, Sakamoto-Hojo ET. Mechanisms underlying the pathophysiology of type 2 diabetes: From risk factors to oxidative stress, metabolic dysfunction, and hyperglycemia. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 874-875:503437. [PMID: 35151421 DOI: 10.1016/j.mrgentox.2021.503437] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/08/2021] [Accepted: 12/12/2021] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes (T2D) is a complex multifactorial disease that emerges from the combination of genetic and environmental factors, and obesity, lifestyle, and aging are the most relevant risk factors. Hyperglycemia is the main metabolic feature of T2D as a consequence of insulin resistance and β-cell dysfunction. Among the cellular alterations induced by hyperglycemia, the overproduction of reactive oxygen species (ROS) and consequently oxidative stress, accompanied by a reduced antioxidant response and impaired DNA repair pathways, represent essential mechanisms underlying the pathophysiology of T2D and the development of late complications. Mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and inflammation are also closely correlated with insulin resistance and β-cell dysfunction. This review focus on the mechanisms by which oxidative stress, mitochondrial dysfunction, ER stress, and inflammation are involved in the pathophysiology of T2D, highlighting the importance of the antioxidant response and DNA repair mechanisms counteracting the development of the disease. Moreover, we indicate evidence on how nutritional interventions effectively improve diabetes care. Additionally, we address key molecular characteristics and signaling pathways shared between T2D and Alzheimer's disease (AD), which might probably be implicated in the risk of T2D patients to develop AD.
Collapse
Affiliation(s)
- Jessica E B F Lima
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Natalia C S Moreira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Elza T Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
17
|
Liang S, Yadav M, Vogel KS, Habib SL. A novel role of snail in regulating tuberin/AMPK pathways to promote renal fibrosis in the new mouse model of type II diabetes. FASEB Bioadv 2021; 3:730-743. [PMID: 34485841 PMCID: PMC8409551 DOI: 10.1096/fba.2020-00134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/25/2021] [Accepted: 04/14/2021] [Indexed: 01/01/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays an important role in tissue fibrosis following chronic exposure to hyperglycemia. This study investigates the role of chronic diabetes in regulating tuberin/snail/AMPK to enhance EMT and increase renal fibrosis. A new mouse model of db/db/TSC2 +/- was generated by backcrossing db/db mice and TSC2 +/- mice. Wild type (WT), db/db, TSC2 +/- and dbdb/TSC2 +/- mice were sacrificed at ages 6 and 8 months old. Tuberin protein level was significantly decreased in kidneys from diabetic compared to WT mice at both ages. In addition, tuberin and E-cadherin protein levels were significantly decreased in dbdb/TSC2 +/- compared to TSC2 +/- and db/db mice. In contrast, p-PS6K, NFkB, snail, vimentin, fibronectin, and α-SMA protein levels were significantly increased in dbdb/TSC2 +/- compared to db/db and TSC2 +/- mice at ages 6 and 8 months. Both downregulation of AMPK by DN-AMPK and downregulation of tuberin by siRNA resulted in increased NFkB, snail, and fibronectin protein expression and decreased E-cadherin protein expression in mouse primary renal proximal tubular cells. Interestingly, downregulation of snail by siRNA increased tuberin expression via feedback through activation of AMPK and reversed the expression of epithelial proteins such as E-cadherin as well as mesenchymal proteins such as fibronectin, NF-KB, vimentin, and α-SMA in mouse primary renal proximal tubular cells isolated from kidneys of four mice genotypes. The data show that chronic diabetes significantly decreases tuberin expression and that provides strong evidence that tuberin is a major key protein involved in regulating EMT. These data also demonstrated a novel role for snail in regulating of AMPK/tuberin to enhance EMT and renal cell fibrosis in diabetes.
Collapse
Affiliation(s)
- Sitai Liang
- Department of Cell Systems and Anatomy The University of Texas Health Science Center San Antonio Texas USA
| | - Mukesh Yadav
- Department of Cell Systems and Anatomy The University of Texas Health Science Center San Antonio Texas USA
| | - Kristine S Vogel
- Department of Cell Systems and Anatomy The University of Texas Health Science Center San Antonio Texas USA
| | - Samy L Habib
- Department of Cell Systems and Anatomy The University of Texas Health Science Center San Antonio Texas USA.,South Texas, Veterans Healthcare System San Antonio Texas USA
| |
Collapse
|
18
|
Nair RG, Vasudev MM, Mavathur R. Role of Yoga and Its Plausible Mechanism in the Mitigation of DNA Damage in Type-2 Diabetes: A Randomized Clinical Trial. Ann Behav Med 2021; 56:235-244. [PMID: 34180500 DOI: 10.1093/abm/kaab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Although yoga is found to be beneficial in the management of type 2 diabetes (T2D), its mechanism of action is poorly understood. T2D is also known to be associated with increased oxidative stress (OS) and DNA damage. PURPOSE This study examines how yoga modulates OS-induced DNA damage and the efficiency of DNA repair in T2D conditions. METHODS In this assessor-masked randomized clinical trial, T2D subjects (n = 61), aged (Mean ± SD, 50.3 ± 4.2) were randomly allocated into Yoga group (31) that received 10 weeks of yoga intervention and Control (30) with routine exercises. Molecular and biochemical assessments were done before and after the intervention period. Structural Equation Modeling using "R" was used for mediation analysis. RESULTS At the end of the 10th week, Yoga group showed significant reduction in DNA damage indicators like Tail Moment (-5.88[95%CI: -10.47 to -1.30]; P = .013) and Olive Tail Moment (-2.93[95%CI: -4.87 to -1.00]; P < .01), oxidative DNA damage marker 8-OHdG (-60.39[95%CI: -92.55 to -28.23]; P < .001) and Fasting Blood Sugar (-22.58[95%CI: -44.33 to -0.83]; P = .042) compared to Control. OGG1 protein expression indicating DNA repair, improved significantly (17.55[95%CI:1.37 to 33.73]; P = .034) whereas Total Antioxidant Capacity did not (5.80[95%CI: -0.86 to 12.47]; P = 0.086). Mediation analysis indicated that improvements in oxidative DNA damage and DNA repair together played a major mediatory role (97.4%) in carrying the effect of yoga. CONCLUSION The beneficial effect of yoga on DNA damage in T2D subjects was found to be mediated by mitigation of oxidative DNA damage and enhancement of DNA repair. CLINICAL TRIAL INFORMATION (www.ctri.nic.in) CTRI/2018/07/014825.
Collapse
Affiliation(s)
- Rajesh G Nair
- Molecular Bioscience Lab, Anvesana Research Labs, S-VYASA, Bangalore, India
| | - Mithila M Vasudev
- Molecular Bioscience Lab, Anvesana Research Labs, S-VYASA, Bangalore, India
| | - Ramesh Mavathur
- Molecular Bioscience Lab, Anvesana Research Labs, S-VYASA, Bangalore, India
| |
Collapse
|
19
|
TP63 Is Significantly Upregulated in Diabetic Kidney. Int J Mol Sci 2021; 22:ijms22084070. [PMID: 33920782 PMCID: PMC8071143 DOI: 10.3390/ijms22084070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 11/17/2022] Open
Abstract
The role of tumor protein 63 (TP63) in regulating insulin receptor substrate 1 (IRS-1) and other downstream signal proteins in diabetes has not been characterized. RNAs extracted from kidneys of diabetic mice (db/db) were sequenced to identify genes that are involved in kidney complications. RNA sequence analysis showed more than 4- to 6-fold increases in TP63 expression in the diabetic mice’s kidneys, compared to wild-type mice at age 10 and 12 months old. In addition, the kidneys from diabetic mice showed significant increases in TP63 mRNA and protein expression compared to WT mice. Mouse proximal tubular cells exposed to high glucose (HG) for 48 h showed significant decreases in IRS-1 expression and increases in TP63, compared to cells grown in normal glucose (NG). When TP63 was downregulated by siRNA, significant increases in IRS-1 and activation of AMP-activated protein kinase (AMPK (p-AMPK-Th172)) occurred under NG and HG conditions. Moreover, activation of AMPK by pretreating the cells with AICAR resulted in significant downregulation of TP63 and increased IRS-1 expression. Ad-cDNA-mediated over-expression of tuberin resulted in significantly decreased TP63 levels and upregulation of IRS-1 expression. Furthermore, TP63 knockdown resulted in increased glucose uptake, whereas IRS-1 knockdown resulted in a decrease in the glucose uptake. Altogether, animal and cell culture data showed a potential role of TP63 as a new candidate gene involved in regulating IRS-1 that may be used as a new therapeutic target to prevent kidney complications in diabetes.
Collapse
|
20
|
Mahmoud HM, Ali AF, Al-Timimi DJ. Relationship Between Zinc Status and DNA Oxidative Damage in Patients with Type 2 Diabetes Mellitus. Biol Trace Elem Res 2021; 199:1276-1279. [PMID: 32666431 DOI: 10.1007/s12011-020-02267-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/21/2020] [Indexed: 12/21/2022]
Abstract
Although zinc deficiency increases the risk of oxidative DNA damage, data regarding the association between zinc and oxidative DNA damage in diabetes are controversial. In this article, we focus on serum zinc levels and its relation to an established biomarker of oxidative DNA damage (8-hydroxy-2-deoxyguanosine) in patients with type 2 diabetes, and to ascertain the beneficial effects of zinc supplementation on the level of oxidative DNA damage. The study consisted of 2 interrelated parts: The first part was a cross-sectional study conducted on patients with type 2 diabetes (n = 297) and healthy individuals (n = 188). The second part was an interventional study that enrolled 38 diabetic patients with low zinc status and high DNA damage. The demographic parameters including age, gender, and body mass index were recorded, and DNA damage marker through 8-hydroxy-2-deoxyguanosine levels, and zinc status of serum zinc, was measured. Significantly higher 8-hydroxy-2-deoxyguanosine levels (P < 0.00) together with lower zinc levels (P < 0.001) were found in the diabetics compared to healthy controls. Patients with low zinc status had higher levels of 8-hydroxy-2-deoxyguanosine compared to patients with normal zinc status. In diabetic patients, a negative correlation of 8-hydroxy-2-deoxyguanosine was observed with zinc (P = 0.070). Zinc supplementation showed a significant decrease in 8-hydroxy-2-deoxyguanosine by (26.0%) and increased in serum zinc by (42.0%). Elevated 8-hydroxy-2-deoxyguanosine levels in conjunction with low zinc status may indicate a high degree of oxidative DNA damage in diabetic patients. The results confirm that zinc supplementation in this group may help correct abnormal levels of 8-hydroxy-2-deoxyguanosine.
Collapse
Affiliation(s)
- Hivi M Mahmoud
- Department of Medical Chemistry, College of Medicine, University of Duhok, Duhok, Kurdistan Region, Iraq.
| | - Ardawan F Ali
- Department of Medical Lab. Technology, Shekhan Technical College of Health, Duhok Polytechnic University, Duhok, Kurdistan Region, Iraq
| | - Dhia J Al-Timimi
- Department of Medical Chemistry, College of Medicine, University of Duhok, Duhok, Kurdistan Region, Iraq
| |
Collapse
|
21
|
Aeeni M, Razi M, Alizadeh A, Alizadeh A. The molecular mechanism behind insulin protective effects on testicular tissue of hyperglycemic rats. Life Sci 2021; 277:119394. [PMID: 33785345 DOI: 10.1016/j.lfs.2021.119394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/21/2021] [Accepted: 03/18/2021] [Indexed: 10/21/2022]
Abstract
AIMS The present study assessed the possible mechanisms by which the insulin regulates the heat shock (HSPs) and transitional proteins expression and consequently ameliorates the oxidative stress-induced damages in germ and sperm cells DNA contents. MAIN METHODS Mature male Wistar rats were distributed into control, Hyperglycemia-induced (HG) and insulin-treated HG-induced (HG-I) groups. Following 8 weeks from HG induction, testicular total antioxidant capacity (TAC), immunoreactivity of 8-oxodG, germ cells mRNA damage, Hsp70-2a, Hsp90, transitional proteins 1 and 2 (TP-1 and -2) mRNA and protein expressions were analyzed. Moreover, the sperm chromatin condensation was assessed by aniline-blue staining, and DNA integrity of germ and sperm cells were analyzed by TUNEL and acrdine-orange staining techniques. KEY FINDINGS The HG animals exhibited significant (p < 0.05) reduction in TAC, HSp70-2a, TP-1 and TP-2 expression levels, and increment in 8-oxodG immunoreactivity, mRNA damage, and Hsp90 expression. However, insulin treatment resulted in (p < 0.05) enhanced TAC level, Hsp70-2a, Hsp90, TP-1 and TP-2 expressions, besides reduced 8-oxodG immunoreactivity and mRNA damage compared to the HG group (p < 0.05). The chromatin condensation and the germ and sperm cells DNA fragmentation were decreased in HG-I group. SIGNIFICANCE Insulin treatment amplifies the testicular TAC level, improves the Hsp70-2a, TP-1, and TP-2 expressions, and boosts the Hsp90-mediated role in DNA repairment process. Consequently, altogether could maintain the HG-induced DNA integrity in the testicular and sperm cells.
Collapse
Affiliation(s)
- Mahsa Aeeni
- Division of Histology & Embryology, Department of Basic Science, Faculty of Veterinary Medicine, P.O.BOX: 1177, Urmia University, Urmia, Iran
| | - Mazdak Razi
- Division of Histology & Embryology, Department of Basic Science, Faculty of Veterinary Medicine, P.O.BOX: 1177, Urmia University, Urmia, Iran.
| | - Alireza Alizadeh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Arash Alizadeh
- Division of Pharmacology and Toxicology, Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
22
|
Fazelzadeh M, Afzalpour ME, Fallah Mohammadi Z, Falah Mohammadi H. The effects of voluntary complex and regular wheel running exercises on the levels of 8-oxoguanine DNA glycosylase, semaphorin 3B, H2O2, and apoptosis in the hippocampus of diabetic rats. Brain Behav 2021; 11:e01988. [PMID: 33471970 PMCID: PMC7994679 DOI: 10.1002/brb3.1988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE One of the most frequent complications associated with diabetes mellitus is apoptosis within the brain which can lead to cognitive disorders. Exercise is considered the best non-pharmacological approach to reduce the severity and extent of cell death through poorly-understood mechanisms. The aim of this study was to investigate the effects of voluntary complex and regular wheel running on the levels of 8-oxoguanine DNA glycosylase (OGG1 ), semaphorin 3B (sema3B), hydrogen peroxide (H2 O2 ), and apoptosis in the hippocampus of diabetic rats. METHODS 48 Wistar male rats were randomly divided into 6 groups: healthy control (C), diabetes control (D), regular wheel running + diabetes (RWD), complex wheel running + diabetes (CWD), healthy regular wheel running (RW), and healthy complex wheel running (CW). The diabetic rat model was produced by intraperitoneal injection of streptozotocin (STZ). The protocol encompassed a 4-week voluntary running training regimen on regular and complex wheel running apparatus. The rats were sacrificed 48 hr after the last training session. To measure the protein concentrations within the hippocampus, ELISA has been utilized. One-way ANOVA was used to compare the groups. RESULTS There were no significant differences in OGG1 protein levels between the groups. H2 O2 level in the D group was significantly higher than the C group (p = .002), while this in RWD and CWD groups was considerably lower than the D group (p = .002 and p = .003, respectively). In the D group, the levels of apoptosis and Sema3B were significantly (p = .001 and p = .007, respectively) higher than C, RWD (p = .001, p = .0001, respectively), and CWD groups (p = .001, p = .006, respectively). Nevertheless, there were not any significant differences between RWD and CWD groups. CONCLUSION The increased levels of Sema3B, H2O2, and apoptosis within the hippocampus associated with diabetes could be noticeably restored by both types of voluntary wheel running protocols.
Collapse
Affiliation(s)
- Mohammad Fazelzadeh
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Birjand, Birjand, Iran
| | | | - Ziya Fallah Mohammadi
- Faculty of Sport Sciences, Department of Exercise Physiology, University of Mazandaran, Babolsar, Iran
| | - Hossein Falah Mohammadi
- Faculty of Natural Sciences, Department of Biology, Ulm University, Baden-Württemberg, Germany
| |
Collapse
|
23
|
Wang W, Ma Y, Huang M, Liang W, Zhao X, Li Q, Wang S, Hu Z, He L, Gao T, Chen J, Pan F, Guo Z. Asymmetrical arginine dimethylation of histone H4 by 8-oxog/OGG1/PRMT1 is essential for oxidative stress-induced transcription activation. Free Radic Biol Med 2021; 164:175-186. [PMID: 33418111 DOI: 10.1016/j.freeradbiomed.2020.12.457] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 01/09/2023]
Abstract
It has been established that 8-oxoguanine DNA glycosylase 1 (OGG1) is the main enzyme removing oxidized guanine under oxidative stress. However, increasing evidence has shown that OGG1 is not only a base excision repair protein but also a new transcriptional coactivator involved in oxidative stress-induced gene expression. Its downstream target genes and the underlying regulatory mechanisms still need to be discerned. Here, it was discovered that c-Myc is a downstream target of OGG1 under oxidative stress and that H4R3me2a is involved in this transcriptional regulation. The increased level of H4R3me2a induced by H2O2 is regulated by OGG1, which may directly interact with the specific arginine methyltransferase PRMT1 and promote the asymmetrical dimethylation of H4R3me1. H4R3me2a enrichment on the promoter of c-Myc can recruit YY1 and activate c-Myc transcription. Moreover, knocking down OGG1 or PRMT1 suppresses c-Myc transcription under oxidative stress by downregulating H4R3me2a formation. Furthermore, the overexpression of wild type (WT) H4R3 promotes c-Myc transcription, but the expression of mutant H4R3Q does not have this effect. Taken together, our data show that the 8-oxoG/OGG1/PRMT1/H4R3me2a/YY1 axis senses oxidative stress and promotes gene transcription.
Collapse
Affiliation(s)
- Wentao Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Ying Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Miaoling Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Weichu Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Xingqi Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Qianwen Li
- Department of Radiotherapy, Taikang Xianlin Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Shiwei Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Tao Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Jinfei Chen
- Department of Radiotherapy, Taikang Xianlin Drum Tower Hospital, Nanjing University, Nanjing, 210000, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China.
| |
Collapse
|
24
|
Pan G, Deshpande M, Pang H, Stemmer PM, Carruthers NJ, Shearn CT, Backos DS, Palaniyandi SS. 4-Hydroxy-2-nonenal attenuates 8-oxoguanine DNA glycosylase 1 activity. J Cell Biochem 2020; 121:4887-4897. [PMID: 32628320 PMCID: PMC7935017 DOI: 10.1002/jcb.29814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/18/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Elevated cellular oxidative stress and oxidative DNA damage are key contributors to impaired cardiac function in diabetes. During chronic inflammation, reactive oxygen species (ROS)-induced lipid peroxidation results in the formation of reactive aldehydes, foremost of which is 4-hydroxy-2-nonenal (4HNE). 4HNE forms covalent adducts with proteins, negatively impacting cellular protein function. During conditions of elevated oxidative stress, oxidative DNA damage such as modification by 8-hydroxydeoxyguanosine (8OHdG) is repaired by 8-oxoguanine glycosylase-1 (OGG-1). Based on these facts, we hypothesized that 4HNE forms adducts with OGG-1 inhibiting its activity, and thus, increases the levels of 8OHG in diabetic heart tissues. To test our hypothesis, we evaluated OGG-1 activity, 8OHG and 4HNE in the hearts of leptin receptor deficient db/db mice, a type-2 diabetic model. We also treated the recombinant OGG-1 with 4HNE to measure direct adduction. We found decreased OGG-1 activity (P > .05), increased 8OHG (P > .05) and increased 4HNE adducts (P > .05) along with low aldehyde dehydrogenase-2 activity (P > .05). The increased colocalization of OGG-1 and 4HNE in cardiomyocytes suggest 4HNE adduction on OGG-1. Furthermore, colocalization of 8OHG and OGG-1 with mitochondrial markers TOM 20 and aconitase, respectively, indicated significant levels of oxidatively-induced mtDNA damage and implicated a role for mitochondrial OGG-1 function. In vitro exposure of recombinant OGG-1 (rOGG-1) with increasing concentrations of 4HNE resulted in a concentration-dependent decrease in OGG-1 activity. Mass spectral analysis of trypsin digests of 4HNE-treated rOGG-1 identified 4HNE adducts on C28, C75, C163, H179, H237, C241, K249, H270, and H282. In silico molecular modeling of 4HNE-K249 OGG-1 and 4HNE-H270 OGG-1 mechanistically supported 4HNE-mediated enzymatic inhibition of OGG-1. In conclusion, these data support the hypothesis that inhibition of OGG-1 by direct modification by 4HNE contributes to decreased OGG-1 activity and increased 8OHG-modified DNA that are present in the diabetic heart.
Collapse
Affiliation(s)
- Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
| | - Mandar Deshpande
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
| | - Haiyan Pang
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences & Proteomics Facility Core, Wayne State University, Detroit, MI, USA, 48201
| | - Nicholas J Carruthers
- Institute of Environmental Health Sciences & Proteomics Facility Core, Wayne State University, Detroit, MI, USA, 48201
| | - Colin T. Shearn
- Department of Pediatrics Division of Pediatric Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80045
| | - Donald S. Backos
- School of Pharmacy, University of Colorado Anschutz Medical Center, Aurora, CO 80045
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202
- Department of Physiology, Wayne State University, Detroit, MI, 48202
| |
Collapse
|
25
|
The role of AMPK in metabolism and its influence on DNA damage repair. Mol Biol Rep 2020; 47:9075-9086. [PMID: 33070285 PMCID: PMC7674386 DOI: 10.1007/s11033-020-05900-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
One of the most complex health disproportions in the human body is the metabolic syndrome (MetS). It can result in serious health consequences such as type 2 diabetes mellitus, atherosclerosis or insulin resistance. The center of energy regulation in human is AMP-activated protein kinase (AMPK), which modulates cells' metabolic pathways and protects them against negative effects of metabolic stress, e.g. reactive oxygen species. Moreover, recent studies show the relationship between the AMPK activity and the regulation of DNA damage repair such as base excision repair (BER) system, which is presented in relation to the influence of MetS on human genome. Hence, AMPK is studied not only in the field of counteracting MetS but also prevention of genetic alterations and cancer development. Through understanding AMPK pathways and its role in cells with damaged DNA it might be possible to improve cell's repair processes and develop new therapies. This review presents AMPK role in eukaryotic cells and focuses on the relationship between AMPK activity and the regulation of BER system through its main component-8-oxoguanine glycosylase (OGG1).
Collapse
|
26
|
Ma Z, Wang W, Wang S, Zhao X, Ma Y, Wu C, Hu Z, He L, Pan F, Guo Z. Symmetrical dimethylation of H4R3: A bridge linking DNA damage and repair upon oxidative stress. Redox Biol 2020; 37:101653. [PMID: 32739156 PMCID: PMC7767741 DOI: 10.1016/j.redox.2020.101653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/27/2020] [Accepted: 07/20/2020] [Indexed: 01/31/2023] Open
Abstract
The DNA lesions caused by oxidative damage are principally repaired by the base excision repair (BER) pathway. 8-oxoguanine DNA glycosylase 1 (OGG1) initiates BER through recognizing and cleaving the oxidatively damaged nucleobase 8-oxo-7,8-dihydroguanine (8-oxoG). How the BER machinery detects and accesses lesions within the context of chromatin is largely unknown. Here, we found that the symmetrical dimethylarginine of histone H4 (producing H4R3me2s) serves as a bridge between DNA damage and subsequent repair. Intracellular H4R3me2s was significantly increased after treatment with the DNA oxidant reagent H2O2, and this increase was regulated by OGG1, which could directly interact with the specific arginine methyltransferase, PRMT5. Arginine-methylated H4R3 could associate with flap endonuclease 1 (FEN1) and enhance its nuclease activity and BER efficiency. Furthermore, cells with a decreased level of H4R3me2s were more susceptible to DNA-damaging agents and accumulated more DNA damage lesions in their genome. Taken together, these results demonstrate that H4R3me2s can be recognized as a reader protein that senses DNA damage and a writer protein that promotes DNA repair.
Collapse
Affiliation(s)
- Zhuang Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China; Institute of DNA Repair Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wentao Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China; Department of Health Technology, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Shiwei Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Xingqi Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Ying Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Congye Wu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, 68, Changle Road, Nanjing, 210006, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Nanjing, 210023, China.
| |
Collapse
|
27
|
Ayinde KS, Olaoba OT, Ibrahim B, Lei D, Lu Q, Yin X, Adelusi TI. AMPK allostery: A therapeutic target for the management/treatment of diabetic nephropathy. Life Sci 2020; 261:118455. [PMID: 32956662 DOI: 10.1016/j.lfs.2020.118455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 12/11/2022]
Abstract
Diabetic nephropathy (DN) is a chronic complication of diabetes mellitus (DM) with approximately 30-40% of patients with DM developing nephropathy, and it is the leading cause of end-stage renal diseases and diabetic morbidity. The pathogenesis of DN is primarily associated with irregularities in the metabolism of glucose and lipid leading to hyperglycemia-induced oxidative stress, which has been a major target together with blood pressure regulation in the control of DN progression. However, the regulation of 5' adenosine monophosphate-activated protein kinase (AMPK), a highly conserved protein kinase for maintaining energy balance and cellular growth and repair has been implicated in the development of DM and its complications. Therefore, targeting AMPK pathway has been explored as a therapeutic strategy for the treatment of diabetes and its complication, although most of the mechanisms have not been fully elucidated. In this review, we discuss the structure of AMPK relevant to understanding its allosteric regulation and its role in the pathogenesis and progression of DN. We also identify therapeutic agents that modulate AMPK and its downstream targets with their specific mechanisms of action in the treatment of DN.
Collapse
Affiliation(s)
| | - Olamide Tosin Olaoba
- Laboratory of Functional and Structural Biochemistry, Federal University of Sao Carlos, Sao Carlos, SP, Brazil
| | - Boyenle Ibrahim
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Du Lei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Temitope Isaac Adelusi
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
28
|
Lan D, Jiang HY, Su X, Zhao Y, Du S, Li Y, Bi R, Zhang DF, Yang Q. Transcriptome-wide Association Study Identifies Genetically Dysregulated Genes in Diabetic Neuropathy. Comb Chem High Throughput Screen 2020; 24:319-325. [PMID: 32772906 DOI: 10.2174/1386207323666200808173745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/03/2020] [Accepted: 07/14/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Complications are the main cause of the disease burden of diabetes. Genes determining the development and progression of diabetic complications remain to be identified. Diabetic neuropathy is the most common and debilitating complication and mainly affects the nerves of legs and feet. In this study, we attempted to identify diabetic neuropathy-specific genes from reliable large-scale genome-wide association studies (GWASs) for diabetes perse. METHODS Taking advantage of publicly available data, we initially converted the GWAS signals to transcriptomic profiles in the tibial nerve using the functional summary-based imputation (FUSION) algorithm. The FUSION-derived genes were then checked to determine whether they were differentially expressed in the sciatic nerve of mouse models of diabetic neuropathy. The dysregulated genes identified in the sciatic nerve were explored in the blood of patients with diabetes. RESULTS We found that eleven out of 452 FUSION-derived genes were regulated by diabetes GWAS loci and were altered in the sciatic nerve of mouse models with early-stage neuropathy. Among the eleven genes, significant (P-value<0.05) expression alterations of HSD17B4, DHX32, MERTK, and SFXN4 could be detected in the blood of human patients. CONCLUSIONS Our analyses identified genes with an effect in the sciatic nerve and provided the possibility of noninvasive early detection of diabetic neuropathy.
Collapse
Affiliation(s)
- Danfeng Lan
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong-Yan Jiang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaoyang Su
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yan Zhao
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Sicheng Du
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Li
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming650223, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming650223, China
| | - Qiuping Yang
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
29
|
Shrikanth CB, Nandini CD. AMPK in microvascular complications of diabetes and the beneficial effects of AMPK activators from plants. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 73:152808. [PMID: 30935723 DOI: 10.1016/j.phymed.2018.12.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/19/2018] [Accepted: 12/23/2018] [Indexed: 05/15/2023]
Abstract
BACKGROUND Diabetes mellitus is a multifactorial disorder with the risk of micro- and macro-vascular complications. High glucose-induced derangements in metabolic pathways are primarily associated with the initiation and progression of secondary complications namely, diabetic nephropathy, neuropathy, and retinopathy. Adenosine monophosphate-activated protein kinase (AMPK) has emerged as an attractive therapeutic target to treat various metabolic disorders including diabetes mellitus. It is a master metabolic regulator that helps in maintaining cellular energy homeostasis by promoting ATP-generating catabolic pathways and inhibiting ATP-consuming anabolic pathways. Numerous pharmacological and plant-derived bioactive compounds that increase AMP-activated protein kinase activation has shown beneficial effects by mitigating secondary complications namely retinopathy, nephropathy, and neuropathy. PURPOSE The purpose of this review is to highlight current knowledge on the role of AMPK and its activators from plant origin in diabetic microvascular complications. METHODS Search engines such as Google Scholar, PubMed, Science Direct and Web of Science are used to extract papers using relevant key words. Papers mainly focusing on the role of AMPK and AMPK activators from plant origin in diabetic nephropathy, retinopathy, and neuropathy was chosen to be highlighted. RESULTS According to results, decrease in AMPK activation during diabetes play a causative role in the pathogenesis of diabetic microvascular complications. Some of the plant-derived bioactive compounds were beneficial in restoring AMPK activity and ameliorating diabetic microvascular complications. CONCLUSION AMPK activators from plant origin are beneficial in mitigating diabetic microvascular complications. These pieces of evidence will be helpful in the development of AMPK-centric therapies to mitigate diabetic microvascular complications.
Collapse
Affiliation(s)
- C B Shrikanth
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka 570 020, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-CFTRI campus, Mysuru, Karnataka 570 020, India
| | - C D Nandini
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, Karnataka 570 020, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-CFTRI campus, Mysuru, Karnataka 570 020, India.
| |
Collapse
|
30
|
Xie X, Chen Y, Liu J, Zhang W, Zhang X, Zha L, Liu W, Ling Y, Li S, Tang S. High glucose induced endothelial cell reactive oxygen species via OGG1/PKC/NADPH oxidase pathway. Life Sci 2020; 256:117886. [PMID: 32497631 DOI: 10.1016/j.lfs.2020.117886] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 01/23/2023]
Abstract
AIMS Reactive oxygen species (ROS) caused by high glucose (HG) is involved in a lot of diseases including diabetes. However, the underlying mechanism of ROS induction by HG remains unclear. Emerging evidence has shown the 8-oxoguanine glycosylase (OGG1) is the main DNA glycosylase responsible for atherosclerosis, obesity, hepatic steatosis, and insulin resistance, and so on. Our aim was to explore the role of OGG1 on HG-mediated endothelial ROS. MAIN METHODS Human umbilical vein endothelial cells (HUVECs) were exposed to HG (30 mM) for different time periods. HG predominantly inhibited OGG1 expression in a time-dependent manner measured by western blotting, qPCR and immunofluorescence. Additionally, HUVECs were cultured with a fluorescent probe, DCFH and DHE, after being subjected to HG. Cell chemiluminescence and flow cytometry results revealed that HG caused endothelial ROS activation. KEY FINDINGS High glucose remarkably decreased endothelial OGG1 expression. The overexpression of OGG1 significantly reversed HG-mediated PKC and NADPH oxidase activities and ROS levels. Moreover, manipulated expression of PKC significantly contacted the role of OGG1 on NADPH oxidase activation. SIGNIFICANCE These results suggest that OGG1 downregulation promoted HG-induced endothelial ROS production and might be a potential clinical treatment target of diabetics.
Collapse
Affiliation(s)
- Xiangrong Xie
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Yan Chen
- Department of Cardiology, Taishan People's Hospital, Taishan, Guangdong 529200, PR China
| | - Jichun Liu
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Wenbo Zhang
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Xuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Lintao Zha
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Wenjie Liu
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Yang Ling
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China
| | - Shu Li
- Department of Pathophysiology, Wannan Medical College, Wuhu, Anhui 241002, PR China.
| | - Shengxing Tang
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, PR China.
| |
Collapse
|
31
|
Pourhanifeh MH, Hosseinzadeh A, Dehdashtian E, Hemati K, Mehrzadi S. Melatonin: new insights on its therapeutic properties in diabetic complications. Diabetol Metab Syndr 2020; 12:30. [PMID: 32280378 PMCID: PMC7140344 DOI: 10.1186/s13098-020-00537-z] [Citation(s) in RCA: 47] [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] [Received: 01/03/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetes and diabetic complications are considered as leading causes of both morbidity and mortality in the world. Unfortunately, routine medical treatments used for affected patients possess undesirable side effects, including kidney and liver damages as well as gastrointestinal adverse reactions. Therefore, exploring the novel therapeutic strategies for diabetic patients is a crucial issue. It has been recently shown that melatonin, as main product of the pineal gland, despite its various pharmacological features including anticancer, anti-aging, antioxidant and anti-inflammatory effects, exerts anti-diabetic properties through regulating various cellular mechanisms. The aim of the present review is to describe potential roles of melatonin in the treatment of diabetes and its complications.
Collapse
Affiliation(s)
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Dehdashtian
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Karim Hemati
- Department of Anesthesiology, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Sabnam S, Rizwan H, Pal S, Pal A. CEES-induced ROS accumulation regulates mitochondrial complications and inflammatory response in keratinocytes. Chem Biol Interact 2020; 321:109031. [PMID: 32142722 DOI: 10.1016/j.cbi.2020.109031] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/13/2020] [Accepted: 03/02/2020] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species (ROS) is mainly produced as a by-product from electron transport chain (ETC) of mitochondria and effectively eliminated by cellular antioxidants. However, 2-chloroethyl ethyl sulfide (CEES) exposure to keratinocytes declined antioxidant capacity and increased accumulation of ROS triggered alteration of mitochondrial activity and apoptosis is lacking. Our findings demonstrated that the electron leakage from the impaired ETC, leading to the accumulation of ROS was gradually elevating with increasing concentration of CEES exposure, which decline the activity of superoxide dismutase (SOD), manganese SOD (MnSOD) and copper-zinc SOD (Cu-ZnSOD) in keratinocytes. Further, excess accumulation of ROS, decreased the mitochondrial membrane potential (ΔΨm) and increased the mitochondrial mass with increasing dose of CEES. CEES exposure provoked the decrease in expression of transcription factor A mitochondrial (TFAM), augmented mitochondrial DNA (mtDNA) damage and altered the mtDNA-encoded oxidative phosphorylation (OXPHOS) subunits. Moreover, fragmented mtDNA translocated into cytosol, where it activated cGAS-STING and interferon regulatory factor3 (IRF3), coinciding with the increased expression of inflammatory mediators and alteration of cell-to-cell communication markers. Pre-treatment of N-acetyl-l-cysteine (NAC) or L-Nω-nitroarginine methyl ester (NAME), hydralazine hydrochloride (Hyd·HCl) or ERK1/2 or phosphoinositide3-kinase (PI3-K)/Akt inhibitors in keratinocyte cells significantly restored the CEES effect. Our findings suggest that CEES-induced mitochondrial ROS production and accumulation leads to mitochondrial dysfunction and inflammatory response in keratinocytes. However, treatment of antioxidants or ERK1/2 or PI3-K/Akt inhibitors is a novel therapeutic option for the keratinocytes complication.
Collapse
Affiliation(s)
- Silpa Sabnam
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India
| | - Huma Rizwan
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India
| | - Sweta Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India
| | - Arttatrana Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India.
| |
Collapse
|
33
|
Urbaniak SK, Boguszewska K, Szewczuk M, Kaźmierczak-Barańska J, Karwowski BT. 8-Oxo-7,8-Dihydro-2'-Deoxyguanosine (8-oxodG) and 8-Hydroxy-2'-Deoxyguanosine (8-OHdG) as a Potential Biomarker for Gestational Diabetes Mellitus (GDM) Development. Molecules 2020; 25:molecules25010202. [PMID: 31947819 PMCID: PMC6982778 DOI: 10.3390/molecules25010202] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/30/2019] [Accepted: 01/01/2020] [Indexed: 12/12/2022] Open
Abstract
The growing clinical and epidemiological significance of gestational diabetes mellitus results from its constantly increasing worldwide prevalence, obesity, and overall unhealthy lifestyle among women of childbearing age. Oxidative stress seems to be the most important predictor of gestational diabetes mellitus development. Disturbances in the cell caused by oxidative stress lead to different changes in biomolecules, including DNA. The nucleobase which is most susceptible to oxidative stress is guanine. Its damage results in two main modifications: 8-hydroxy-2′-deoxyguanosineor 8-oxo-7,8-dihydro-2′-deoxyguanosine. Their significant level can indicate pathological processes during pregnancy, like gestational diabetes mellitus and probably, type 2 diabetes mellitus after pregnancy. This review provides an overview of current knowledge on the use of 8-hydroxy-2′-deoxyguanosineand/or 8-oxo-7,8-dihydro-2′-deoxyguanosine as a biomarker in gestational diabetes mellitus and allows us to understand the mechanism of 8-hydroxy-2′-deoxyguanosineand/or 8-oxo-7,8-dihydro-2′-deoxyguanosine generation during this disease.
Collapse
|
34
|
Ramteke P, Deb A, Shepal V, Bhat MK. Hyperglycemia Associated Metabolic and Molecular Alterations in Cancer Risk, Progression, Treatment, and Mortality. Cancers (Basel) 2019; 11:E1402. [PMID: 31546918 PMCID: PMC6770430 DOI: 10.3390/cancers11091402] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer and diabetes are amongst the leading causes of deaths worldwide. There is an alarming rise in cancer incidences and mortality, with approximately 18.1 million new cases and 9.6 million deaths in 2018. A major contributory but neglected factor for risk of neoplastic transformation is hyperglycemia. Epidemiologically too, lifestyle patterns resulting in high blood glucose level, with or without the role of insulin, are more often correlated with cancer risk, progression, and mortality. The two conditions recurrently exist in comorbidity, and their interplay has rendered treatment regimens more challenging by restricting the choice of drugs, affecting surgical consequences, and having associated fatal complications. Limited comprehensive literature is available on their correlation, and a lack of clarity in understanding in such comorbid conditions contributes to higher mortality rates. Hence, a critical analysis of the elements responsible for enhanced mortality due to hyperglycemia-cancer concomitance is warranted. Given the lifestyle changes in the human population, increasing metabolic disorders, and glucose addiction of cancer cells, hyperglycemia related complications in cancer underline the necessity for further in-depth investigations. This review, therefore, attempts to shed light upon hyperglycemia associated factors in the risk, progression, mortality, and treatment of cancer to highlight important mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Pranay Ramteke
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune-411 007, India.
| | - Ankita Deb
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune-411 007, India.
| | - Varsha Shepal
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune-411 007, India.
| | - Manoj Kumar Bhat
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune-411 007, India.
| |
Collapse
|
35
|
Obesity, DNA Damage, and Development of Obesity-Related Diseases. Int J Mol Sci 2019; 20:ijms20051146. [PMID: 30845725 PMCID: PMC6429223 DOI: 10.3390/ijms20051146] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity has been recognized to increase the risk of such diseases as cardiovascular diseases, diabetes, and cancer. It indicates that obesity can impact genome stability. Oxidative stress and inflammation, commonly occurring in obesity, can induce DNA damage and inhibit DNA repair mechanisms. Accumulation of DNA damage can lead to an enhanced mutation rate and can alter gene expression resulting in disturbances in cell metabolism. Obesity-associated DNA damage can promote cancer growth by favoring cancer cell proliferation and migration, and resistance to apoptosis. Estimation of the DNA damage and/or disturbances in DNA repair could be potentially useful in the risk assessment and prevention of obesity-associated metabolic disorders as well as cancers. DNA damage in people with obesity appears to be reversible and both weight loss and improvement of dietary habits and diet composition can affect genome stability.
Collapse
|
36
|
Djelić N, Radaković M, Borozan S, Dimirijević-Srećković V, Pajović N, Vejnović B, Borozan N, Bankoglu EE, Stopper H, Stanimirović Z. Oxidative stress and DNA damage in peripheral blood mononuclear cells from normal, obese, prediabetic and diabetic persons exposed to adrenaline in vitro. Mutat Res 2019; 843:81-89. [PMID: 31421743 DOI: 10.1016/j.mrgentox.2019.01.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 01/08/2023]
Abstract
Diabetes represents one of the major health concerns, especially in developed countries. Some hormones such as the stress hormone adrenaline can induce reactive oxygen species (ROS) and may worsen the diabetes. Therefore, the main aim of the investigation was to find out whether peripheral blood mononuclear cells (PBMCs) from normal persons have less DNA damage induced by adrenaline (0.1, 1 and 10 μM) in comparison to PBMCs from obese, prediabetic and diabetic patients. Also, the biochemical parameters of oxidative stress (TBARS, catalase) and lactate dehydrogenase were monitored. It was observed that higher concentrations of adrenaline (1 and 10 μM) induced DNA damage in the obese, prediabetic and diabetic groups. In healthy individuals only the highest concentration of adrenaline caused significant increase in the DNA damage. In summary, total comet score (TCS) comparison has shown significant differences between groups, and DNA damaging effects of adrenaline were most evident in diabetic patients. The results of the biochemical analysis also demonstrate that adrenaline exerts most obvious effects in diabetic individuals which is manifested as significant change of parameters of oxidative stress. In summary, the obtained results demonstrated that diabetics are more sensitive to genotoxic effects of adrenaline and this effect probably resulted from decreased antioxidative defence mechanisms in various stages of progression through diabetes. Therefore, these results could contribute to a better understanding of a role of endocrine factors to damage of cellular biomolecules which could be useful in finding novel therapeutic approaches and lifestyle changes with an aim to lower the possibility of diabetes complications.
Collapse
Affiliation(s)
- Ninoslav Djelić
- Department of Biology, Faculty of Veterinary Medicine, University of Belgrade, Serbia.
| | - Milena Radaković
- Department of Biology, Faculty of Veterinary Medicine, University of Belgrade, Serbia.
| | - Sunčica Borozan
- Department of Chemistry, Faculty of Veterinary Medicine, University of Belgrade, Serbia.
| | | | - Nevena Pajović
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Belgrade, Serbia.
| | - Branislav Vejnović
- Department of Economics and Statistics, Faculty of Veterinary Medicine, University of Belgrade, Serbia.
| | | | - Ezgi Eylül Bankoglu
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany.
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany.
| | - Zoran Stanimirović
- Department of Biology, Faculty of Veterinary Medicine, University of Belgrade, Serbia.
| |
Collapse
|
37
|
Davargaon RS, Sambe AD, Muthangi V V S. Toxic effect of high glucose on cardiomyocytes, H9c2 cells: Induction of oxidative stress and ameliorative effect of trolox. J Biochem Mol Toxicol 2018; 33:e22272. [PMID: 30512247 DOI: 10.1002/jbt.22272] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/11/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022]
Abstract
Oxidative stress (OS) has been implicated in a variety of pathological conditions, including diabetes mellitus, characterized by hyperglycemia. In the present study, OS induced by hyperglycemia and the effect of trolox, a vitamin E analog, were studied in cardiomyocytes and H9c2 cells exposed to 15 to 33 mM glucose (HG) for 24 to 72 hours in Dulbecco modified Eagle medium. Cells treated wirh 24 or 33 mM glucose for 24 hours or above showed decreased viability and adenosine triphosphate (ATP) content with a concomitant increase in radicals of oxygen species, calcium (Ca2+ ), mitochondrial permeability transition, and oxidative markers, confirming that the cells were under stress. However, upon exposure to 15 mM glucose for 24 hours, H9c2 cells maintained homeostasis and ATP generation. Pretreatment of cells with trolox reduced HG-induced OS to control levels. Here, we report that the toxic effect of HG is highly regulated and that OS induction can be prevented with Trolox, a potential inhibitor of membrane damage.
Collapse
Affiliation(s)
| | - Asha Devi Sambe
- Department of Zoology, Laboratory of Gerontology, J.B. Campus, Bangalore University, Bangalore, India
| | | |
Collapse
|
38
|
Huy H, Song HY, Kim MJ, Kim WS, Kim DO, Byun JE, Lee J, Park YJ, Kim TD, Yoon SR, Choi EJ, Lee CH, Noh JY, Jung H, Choi I. TXNIP regulates AKT-mediated cellular senescence by direct interaction under glucose-mediated metabolic stress. Aging Cell 2018; 17:e12836. [PMID: 30168649 PMCID: PMC6260918 DOI: 10.1111/acel.12836] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 07/16/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
Aging is associated with an inevitable and universal loss of cell homeostasis and restricts an organism's lifespan by an increased susceptibility to diseases and tissue degeneration. The glucose uptake associated with producing energy for cell survival is one of the major causes of ROS production under physiological conditions. However, the overall mechanisms by which glucose uptake results in cellular senescence remain mysterious. In this study, we found that TXNIP deficiency accelerated the senescent phenotypes of MEF cells under high glucose condition. TXNIP‐/‐ MEF cells showed greater induced glucose uptake and ROS levels than wild‐type cells, and N‐acetylcysteine (NAC) treatment rescued the cellular senescence of TXNIP‐/‐ MEF cells. Interestingly, TXNIP‐/‐ MEF cells showed continuous activation of AKT during long‐term subculture, and AKT signaling inhibition completely blocked the cellular senescence of TXNIP‐/‐ MEF cells. In addition, we found that TXNIP interacted with AKT via the PH domain of AKT, and their interaction was increased by high glucose or H2O2 treatment. The inhibition of AKT activity by TXNIP was confirmed using western blotting and an in vitro kinase assay. TXNIP deficiency in type 1 diabetes mice (Akita) efficiently decreased the blood glucose levels and finally increased mouse survival. However, in normal mice, TXNIP deficiency induced metabolic aging of mice and cellular senescence of kidney cells by inducing AKT activity and aging‐associated gene expression. Altogether, these results suggest that TXNIP regulates cellular senescence by inhibiting AKT pathways via a direct interaction under conditions of glucose‐derived metabolic stress.
Collapse
Affiliation(s)
- Hangsak Huy
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Functional Genomics; University of Science and Technology (UST); Daejeon Korea
| | - Hae Young Song
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Mi Jeong Kim
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Won Sam Kim
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Dong Oh Kim
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Jae-Eun Byun
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Biochemistry, School of Life Sciences; Chungbuk National University; Cheongju Korea
| | - Jungwoon Lee
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Young-Jun Park
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Functional Genomics; University of Science and Technology (UST); Daejeon Korea
| | - Tae-Don Kim
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Functional Genomics; University of Science and Technology (UST); Daejeon Korea
| | - Suk Ran Yoon
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Functional Genomics; University of Science and Technology (UST); Daejeon Korea
| | - Eun-Ji Choi
- Department of Hematology, Asan Medical Center; University of Ulsan College of Medicine; Seoul Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Ji-Yoon Noh
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
| | - Haiyoung Jung
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Functional Genomics; University of Science and Technology (UST); Daejeon Korea
| | - Inpyo Choi
- Immunotherapy Convergence Research Center; Korea Research Institute of Bioscience and Biotechnology (KRIBB); Daejeon Korea
- Department of Functional Genomics; University of Science and Technology (UST); Daejeon Korea
| |
Collapse
|
39
|
Liang S, Medina EA, Li B, Habib SL. Preclinical evidence of the enhanced effectiveness of combined rapamycin and AICAR in reducing kidney cancer. Mol Oncol 2018; 12:1917-1934. [PMID: 30107094 PMCID: PMC6210038 DOI: 10.1002/1878-0261.12370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/19/2018] [Accepted: 07/26/2018] [Indexed: 12/31/2022] Open
Abstract
Loss of Von Hippel-Lindau in renal carcinoma cells results in upregulation of the activity of hypoxia-inducible factor (HIF-α), a major transcription factor involved in kidney cancer. Rapamycin as mammalian target of rapamycin inhibitor and 5-aminoimidazole-4-carboxamide-riboside (AICAR) as AMPK activator are used separately to treat cancer patients. In the current study, the possible additive effect of drug combinations in reducing kidney tumorigenesis was investigated. Treatment with drug combinations significantly decreased cell proliferation, increased cell apoptosis, and abolished Akt phosphorylation and HIF-2α expression in renal cell carcinoma cells, including primary cells isolated from kidney cancer patients. Significant decreases in cell migration and invasion were detected using drug combinations. Drug combinations effectively abolished binding of HIF-2α to the Akt promoter and effected formation of the DNA-protein complex in nuclear extracts from 786-O cells, as demonstrated using electromobility shift assay and examination of Akt promoter activity. Importantly, we tested the effect of each drug and the combined drugs on kidney tumor size in the nude mouse model. Our data show that treatment with rapamycin, AICAR, and rapamycin+AICAR decreased tumor size by 38%, 36%, and 80%, respectively, suggesting that drug combinations have an additive effect in reducing tumor size compared with use of each drug alone. Drug combinations effectively decreased cell proliferation, increased apoptotic cells, and significantly decreased p-Akt, HIF-2α, and vascular endothelial growth factor expression in tumor kidney tissues from mice. These results show for the first time that drug combinations are more effective than single drugs in reducing kidney tumor progression. This study provides important evidence that may lead to the initiation of pre-clinical trials in patients with kidney cancer.
Collapse
Affiliation(s)
- Sitai Liang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, USA
| | - Edward A Medina
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, TX, USA
| | - Boajie Li
- Bio-X Institutes, Shanghai Jiao Tong University, China
| | - Samy L Habib
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX, USA.,South Texas Veterans Health Care System, San Antonio, TX, USA
| |
Collapse
|
40
|
Giri B, Dey S, Das T, Sarkar M, Banerjee J, Dash SK. Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity. Biomed Pharmacother 2018; 107:306-328. [PMID: 30098549 DOI: 10.1016/j.biopha.2018.07.157] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/15/2018] [Accepted: 07/31/2018] [Indexed: 02/09/2023] Open
Abstract
Chronic exposure of glucose rich environment creates several physiological and pathophysiological changes. There are several pathways by which hyperglycemia exacerbate its toxic effect on cells, tissues and organ systems. Hyperglycemia can induce oxidative stress, upsurge polyol pathway, activate protein kinase C (PKC), enhance hexosamine biosynthetic pathway (HBP), promote the formation of advanced glycation end-products (AGEs) and finally alters gene expressions. Prolonged hyperglycemic condition leads to severe diabetic condition by damaging the pancreatic β-cell and inducing insulin resistance. Numerous complications have been associated with diabetes, thus it has become a major health issue in the 21st century and has received serious attention. Dysregulation in the cardiovascular and reproductive systems along with nephropathy, retinopathy, neuropathy, diabetic foot ulcer may arise in the advanced stages of diabetes. High glucose level also encourages proliferation of cancer cells, development of osteoarthritis and potentiates a suitable environment for infections. This review culminates how elevated glucose level carries out its toxicity in cells, metabolic distortion along with organ dysfunction and elucidates the complications associated with chronic hyperglycemia.
Collapse
Affiliation(s)
- Biplab Giri
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India.
| | - Sananda Dey
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Tanaya Das
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Mrinmoy Sarkar
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India.
| |
Collapse
|
41
|
Pavlou S, Lindsay J, Ingram R, Xu H, Chen M. Sustained high glucose exposure sensitizes macrophage responses to cytokine stimuli but reduces their phagocytic activity. BMC Immunol 2018; 19:24. [PMID: 29996768 PMCID: PMC6042333 DOI: 10.1186/s12865-018-0261-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/04/2018] [Indexed: 01/06/2023] Open
Abstract
Background Macrophages are tissue resident immune cells important for host defence and homeostasis. During diabetes, macrophages and other innate immune cells are known to have a pro-inflammatory phenotype, which is believed to contribute to the pathogenesis of various diabetic complications. However, diabetic patients are highly susceptible to bacterial infections, and often have impaired wound healing. The molecular mechanism underlying the paradox of macrophage function in diabetes is not fully understood. Recent evidence suggests that macrophage functions are governed by metabolic reprograming. Diabetes is a disorder that affects glucose metabolism; dysregulated macrophage function in diabetes may be related to alterations in their metabolic pathways. In this study, we seek to understand the effect of high glucose exposure on macrophage phenotype and functions. Results Bone marrow cells were cultured in short or long term high glucose and normal glucose medium; the number and phenotype of bone marrow derived macrophages were not affected by long-term high glucose treatment. Short-term high glucose increased the expression of IL-1β. Long-term high glucose increased the expression of IL-1β and TNFα but reduced the expression of IL-12p40 and nitric oxide production in M1 macrophage. The treatment also increased Arg-1 and IL-10 expression in M2 macrophages. Phagocytosis and bactericidal activity was reduced in long-term high glucose treated macrophages and peritoneal macrophages from diabetic mice. Long-term high glucose treatment reduced macrophage glycolytic capacity and glycolytic reserve without affecting mitochondrial ATP production and oxidative respiration. Conclusion Long-term high glucose sensitizes macrophages to cytokine stimulation and reduces phagocytosis and nitric oxide production, which may be related to impaired glycolytic capacity. Electronic supplementary material The online version of this article (10.1186/s12865-018-0261-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofia Pavlou
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Jaime Lindsay
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Rebecca Ingram
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
42
|
Al-Obaidi N, Mohan S, Liang S, Zhao Z, Nayak BK, Li B, Sriramarao P, Habib SL. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes. FASEB J 2018; 33:373-387. [PMID: 29975570 DOI: 10.1096/fj.201800555rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Chronic exposure of tubular renal cells to high glucose contributes to tubulointerstitial changes in diabetic nephropathy. In the present study, we identified a new fibrosis gene called galectin-1 (Gal-1), which is highly expressed in tubular cells of kidneys of type 1 and type 2 diabetic mouse models. Gal-1 protein and mRNA expression showed significant increase in kidney cortex of heterozygous Akita+/- and db/db mice compared with wild-type mice. Mouse proximal tubular cells exposed to high glucose showed significant increase in phosphorylation of Akt and Gal-1. We cloned Gal-1 promoter and identified the transcription factor AP4 as binding to the Gal-1 promoter to up-regulate its function. Transfection of cells with plasmid carrying mutations in the binding sites of AP4 to Gal-1 promoter resulted in decreased protein function of Gal-1. In addition, inhibition of Gal-1 by OTX-008 showed significant decrease in p-Akt/AP4 and protein-promoter activity of Gal-1 and fibronectin. Moreover, down-regulation of AP4 by small interfering RNA resulted in a significant decrease in protein expression and promoter activity of Gal-1. We found that kidney of Gal-1-/- mice express very low levels of fibronectin protein. In summary, Gal-1 is highly expressed in kidneys of type 1 and 2 diabetic mice, and AP4 is a major transcription factor that activates Gal-1 under hyperglycemia. Inhibition of Gal-1 by OTX-008 blocks activation of Akt and prevents accumulation of Gal-1, suggesting a novel role of Gal-1 inhibitor as a possible therapeutic target to treat renal fibrosis in diabetes.-Al-Obaidi, N., Mohan, S., Liang, S., Zhao, Z., Nayak, B. K., Li, B., Sriramarao, P., Habib, S. L. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Noor Al-Obaidi
- Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, Texas, USA
| | - Sumathy Mohan
- Department of Pathology, University of Texas Health, San Antonio, Texas, USA
| | - Sitai Liang
- Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, Texas, USA
| | - Zhenze Zhao
- Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, Texas, USA
| | - Bijaya K Nayak
- Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, Texas, USA
| | - Boajie Li
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - P Sriramarao
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA; and
| | - Samy L Habib
- Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, Texas, USA.,Geriatric Research Education and Clinical Center, South Texas, Veterans Healthcare System, San Antonio, Texas, USA
| |
Collapse
|
43
|
Tozour JN, Delahaye F, Suzuki M, Praiss A, Zhao Y, Cai L, Heo HJ, Greally JM, Hughes F. Intrauterine Hyperglycemia Is Associated with an Impaired Postnatal Response to Oxidative Damage. Stem Cells Dev 2018; 27:683-691. [PMID: 29598691 DOI: 10.1089/scd.2017.0232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hyperglycemia and other adverse exposures early in life that reprogram stem cells may lead to long-lasting phenotypic influences over the lifetime of an individual. Hyperglycemia and oxidative stress cause DNA damage when they exceed the protective capabilities of the cell, in turn affecting cellular function. DNA damage in response to hyperglycemia and oxidative stress was studied in human umbilical cord mesenchymal stem cells (hUC-MSCs) from large-for-gestational-age (LGA) infants of mothers with gestational diabetes mellitus (LGA-GDM) and control subjects. We tested the response of these cells to hyperglycemia and oxidative stress, measuring reactive oxygen species (ROS) levels and antioxidant enzyme activities. We find that hUC-MSCs from LGA-GDM infants have increased DNA damage when exposed to oxidative stress. With the addition of hyperglycemic conditions, these cells have an increase in ROS and a decrease in antioxidant glutathione peroxidase (GPx) activity, indicating a mechanism for the increased ROS and DNA damage. This study demonstrates that a memory of in utero hyperglycemia, mediated through downregulation of GPx activity, leads to an increased susceptibility to oxidative stress. The alteration of GPx function in self-renewing stem cells, can mediate the effect of intrauterine hyperglycemia to be propagated into adulthood and contribute to disease susceptibility.
Collapse
Affiliation(s)
- Jessica N Tozour
- 1 Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine , Bronx, New York
| | - Fabien Delahaye
- 1 Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine , Bronx, New York.,2 Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine , Bronx, New York
| | - Masako Suzuki
- 1 Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine , Bronx, New York
| | - Aaron Praiss
- 2 Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine , Bronx, New York
| | - Yongmei Zhao
- 2 Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine , Bronx, New York
| | - Lingguang Cai
- 2 Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine , Bronx, New York
| | - Hye J Heo
- 3 Department of Obstetrics and Gynecology, NYU Winthrop Hospital , Mineola, New York
| | - John M Greally
- 1 Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine , Bronx, New York.,4 Department of Pediatrics (Genetics), Albert Einstein College of Medicine , Bronx, New York
| | - Francine Hughes
- 5 Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, NYU Langone Medical Center , New York, New York
| |
Collapse
|
44
|
Sanchez M, Roussel R, Hadjadj S, Moutairou A, Marre M, Velho G, Mohammedi K. Plasma concentrations of 8-hydroxy-2'-deoxyguanosine and risk of kidney disease and death in individuals with type 1 diabetes. Diabetologia 2018; 61:977-984. [PMID: 29185011 DOI: 10.1007/s00125-017-4510-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Oxidative stress is involved in the pathogenesis of diabetic kidney disease. We evaluated the association between 8-hydroxy-2'-deoxyguanosine (8-OHdG), a marker of DNA oxidative damage, and end-stage renal disease (ESRD) or death in individuals with type 1 diabetes. METHODS Plasma 8-OHdG concentrations were measured at baseline in participants with type 1 diabetes from GENEDIAB (n = 348) and GENESIS (n = 571) cohorts. A follow-up was conducted in 205 and 499 participants for a mean ± SD duration of 8.9 ± 2.3 years and 5.2 ± 1.9 years, respectively. We tested associations between 8-OHdG concentrations and urinary albumin concentration (UAC) or eGFR at baseline, and the risk of ESRD or all-cause mortality during follow-up. Analyses were performed in pooled cohorts. RESULTS The highest UAC (geometric mean [95% CI]) was observed in the third 8-OHdG tertile (tertile 1, 9 [6, 13] mg/l; tertile 2, 10 [7, 16] mg/l; tertile 3, 16 [10, 25] mg/l; p = 0.36 for tertile 1 vs tertile 2 and p = 0.003 for tertile 3 vs tertile 1) after adjustment for potential confounding covariates. The lowest eGFR (mean [95% CI]) was observed in the third tertile (tertile 1, 87 [82, 93] ml min-1 1.73 m-2; tertile 2, 88 [82, 94] ml min-1 1.73 m-2; tertile 3, 74 [68, 80] ml min-1 1.73 m-2; p = 0.61 for tertile 1 vs tertile 2; p < 0.001 for tertile 3 vs tertile 1). ESRD and death occurred in 48 and 64 individuals, respectively. The HR for ESRD, but not death, was higher in the third tertile than in the first (tertile 2 vs tertile 1, 1.45 [0.45, 5.04], p = 0.54; tertile 3 vs tertile 1, 3.05 [1.16, 9.60], p = 0.02) after multiple adjustments. CONCLUSIONS/INTERPRETATION Higher plasma concentrations of 8-OHdG were independently associated with increased risk of kidney disease in individuals with type 1 diabetes, suggesting that this marker can be used to evaluate the progression of diabetic kidney disease.
Collapse
Affiliation(s)
- Manuel Sanchez
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Department of Geriatric Medicine, Assistance Publique Hôpitaux de Paris, Bichat Hospital, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Ronan Roussel
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, Assistance Publique Hôpitaux de Paris, Bichat Hospital, DHU FIRE, 46 rue Henri Huchard, 75877, Paris Cedex 18, France
| | - Samy Hadjadj
- Department of Endocrinology and Diabetology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
- Inserm, Research Unit 1082, Poitiers, France
- Université de Poitiers, UFR de Médecine et Pharmacie, Poitiers, France
| | - Abdul Moutairou
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Michel Marre
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Department of Diabetology, Endocrinology and Nutrition, Assistance Publique Hôpitaux de Paris, Bichat Hospital, DHU FIRE, 46 rue Henri Huchard, 75877, Paris Cedex 18, France
| | - Gilberto Velho
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Kamel Mohammedi
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.
- Department of Diabetology, Endocrinology and Nutrition, Assistance Publique Hôpitaux de Paris, Bichat Hospital, DHU FIRE, 46 rue Henri Huchard, 75877, Paris Cedex 18, France.
| |
Collapse
|
45
|
Kurosaki Y, Imoto A, Kawakami F, Yokoba M, Takenaka T, Ichikawa T, Katagiri M, Ishii N. Oxidative stress increases megalin expression in the renal proximal tubules during the normoalbuminuric stage of diabetes mellitus. Am J Physiol Renal Physiol 2018; 314:F462-F470. [DOI: 10.1152/ajprenal.00108.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Megalin, an endocytic receptor expressed in proximal tubule cells, plays a critical role in renal tubular protein reabsorption and is associated with the albuminuria observed in diabetic nephropathy. We have previously reported increased oxidant production in the renal cortex during the normoalbuminuric stage of diabetes mellitus (DM); however, the relationship between oxidative stress and renal megalin expression during the normoalbuminuric stage of DM remains unclear. In the present study, we evaluated whether oxidative stress affects megalin expression in the normoalbuminuric stage of DM in a streptozotocin-induced diabetic rat model and in immortalized human proximal tubular cells (HK-2). We demonstrated that increased expression of renal megalin accompanies oxidative stress during the early stage of DM, before albuminuria development. Telmisartan treatment prevented the diabetes-induced elevation in megalin level, possibly through an oxidative stress-dependent mechanism. In HK-2 cells, hydrogen peroxide significantly increased megalin levels in a dose- and time-dependent manner; however, the elevation in megalin expression was decreased following prolonged exposure to severe oxidative stress induced by 0.4 mmol/l hydrogen peroxide. High-glucose treatment also significantly increased megalin expression in HK-2 cells. Concurrent administration of the antioxidant N-acetyl-cysteine blocked the effects of high glucose on megalin expression. Furthermore, the hydrogen peroxide-induced increase in megalin expression was blocked by treatment with phosphatidylinositol 3-kinase and Akt inhibitors. Increase of phosphorylated Akt expression was also seen in the renal cortex of diabetic rats. Taken together, our results indicate that mild oxidative stress increases renal megalin expression through the phosphatidylinositol 3-kinase-Akt pathway in the normoalbuminuric stage of DM.
Collapse
Affiliation(s)
- Yoshifumi Kurosaki
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Akemi Imoto
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Fumitaka Kawakami
- Department of Pathological Biochemistry, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Masanori Yokoba
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Tsuneo Takenaka
- Department of Medicine, International University of Health and Welfare, Tokyo, Japan
| | - Takafumi Ichikawa
- Department of Pathological Biochemistry, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Masato Katagiri
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Naohito Ishii
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| |
Collapse
|
46
|
Abstract
The mechanism by which TSC2 inactivation or deficiency contributes to the pathology of tuberous sclerosis complex (TSC) is not fully clear. We show that renal angiomyolipomas from TSC patients and kidney cortex from Tsc2+/− mice exhibit elevated levels of reactive oxygen species (ROS). Downregulation of tuberin (protein encoded by TSC2 gene) in renal proximal tubular epithelial cells significantly increased ROS concomitant with enhanced Nox4. Similarly, we found elevated levels of Nox4 in the renal cortex of Tsc2+/− mice and in the renal angiomyolipomas from TSC patients. Tuberin deficiency is associated with activation of mTORC1. Rapamycin, shRNAs targeting raptor, or inhibition of S6 kinase significantly inhibited the expression of Nox4, resulting in attenuation of production of ROS in tuberin-downregulated proximal tubular epithelial cells. In contrast, activation of mTORC1 increased Nox4 and ROS. These results indicate that Nox4 may be a potential target for tuberin-deficiency-derived diseases. Using a xenograft model from tuberin-null tubular cells in nude mice, both anti-sense Nox4 and GKT137831, a specific inhibitor of Nox1/4, significantly inhibited the tumor growth. Thus, our results demonstrate the presence of an antagonistic relationship between tuberin and Nox4 to drive oncogenesis in the tuberin deficiency syndrome and identify Nox4 as a target to develop a therapy for TSC.
Collapse
|
47
|
Sifuentes-Franco S, Padilla-Tejeda DE, Carrillo-Ibarra S, Miranda-Díaz AG. Oxidative Stress, Apoptosis, and Mitochondrial Function in Diabetic Nephropathy. Int J Endocrinol 2018; 2018:1875870. [PMID: 29808088 PMCID: PMC5902001 DOI: 10.1155/2018/1875870] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/07/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetic nephropathy (DN) is the second most frequent and prevalent complication of diabetes mellitus (DM). The increase in the production of oxidative stress (OS) is induced by the persistent hyperglycemic state capable of producing oxidative damage to the macromolecules (lipids, carbohydrates, proteins, and nucleic acids). OS favors the production of oxidative damage to the histones of the double-chain DNA and affects expression of the DNA repairer enzyme which leads to cell death from apoptosis. The chronic hyperglycemic state unchains an increase in advanced glycation end-products (AGE) that interact through the cellular receptors to favor activation of the transcription factor NF-κB and the protein kinase C (PKC) system, leading to the appearance of inflammation, growth, and augmentation of synthesis of the extracellular matrix (ECM) in DN. The reactive oxygen species (ROS) play an important role in the pathogenesis of diabetic complications because the production of ROS increases during the persistent hyperglycemia. The primary source of the excessive production of ROS is the mitochondria with the capacity to exceed production of endogenous antioxidants. Due to the fact that the mechanisms involved in the development of DN have not been fully clarified, there are different approaches to specific therapeutic targets or adjuvant management alternatives in the control of glycemia in DN.
Collapse
Affiliation(s)
- Sonia Sifuentes-Franco
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, University of Guadalajara, Guadalajara, JAL, Mexico
| | - Diego Enrique Padilla-Tejeda
- Programa de Químico Farmacéutico Biotecnologo, Escuela de Ciencias de la Salud, Campus Zapopan, Universidad del Valle de México, Guadalajara, JAL, Mexico
| | - Sandra Carrillo-Ibarra
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, University of Guadalajara, Guadalajara, JAL, Mexico
| | - Alejandra Guillermina Miranda-Díaz
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, University of Guadalajara, Guadalajara, JAL, Mexico
| |
Collapse
|
48
|
Ercan S, Şahin P, Kencebay C, Derin N, Çelik Özenci Ç. Evaluation of mTOR signaling pathway proteins in rat gastric mucosa exposed to sulfite and ghrelin. TURKISH JOURNAL OF GASTROENTEROLOGY 2017; 29:94-100. [PMID: 29082888 DOI: 10.5152/tjg.2017.17294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND/AIMS Mammalian target of rapamycin (mTOR) signaling serves as a central regulator of cell growth, proliferation, and survival. In this study, we planned to evaluate the expressions of mTOR signaling constituents (p-p70S6K, p-mTOR, and p-Tuberin) in rat gastric mucosa and to compare the results in sulfite- and sulfite+ghrelin-exposed groups. MATERIALS AND METHODS Rats were divided into three groups: the control group (C), the sodium metabisulfite (Na2S2O5) (S) group, and sulfite+ghrelin (SG) group. Sodium metabisulfite at 100 mg/kg/day was administered via gavage, and ghrelin at 20 μg/kg/day was administered intraperitoneally for 35 days. We have used immunohistochemistry for mTOR signaling pathway components. RESULTS There were no significant differences for p-p70S6K and p-mTOR expression among the C, S, and SG groups. Tuberin expression was significantly increased in the S group compared to the C group. Furthermore, tuberin expression was found to be significantly decreased in the SG group. CONCLUSION This study is the first one in the literature that shows the expression of mTOR signaling proteins in gastric mucosa of rats exposed to sulfite and ghrelin. Furthermore, it demonstrates that ghrelin treatment reduces p-Tuberin expression induced by ingested sulfite.
Collapse
Affiliation(s)
- Sevim Ercan
- Department of Medical Services and Techniques, Akdeniz University Vocational School of Health Services, Antalya, Turkey
| | - Pınar Şahin
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Ceren Kencebay
- Department of Biophysics, Akdeniz University School of Medicine, Antalya, Turkey
| | - Narin Derin
- Department of Biophysics, Akdeniz University School of Medicine, Antalya, Turkey
| | - Çiler Çelik Özenci
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
49
|
Eskelinen TJ, Kotsar A, Tammela TL, Murtola TJ. Components of metabolic syndrome and prognosis of renal cell cancer. Scand J Urol 2017; 51:435-441. [DOI: 10.1080/21681805.2017.1352616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | - Andres Kotsar
- Department of Urology, Tarto University Hospital, Tarto, Estonia
| | - Teuvo L.J. Tammela
- School of Medicine, University of Tampere, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Teemu J. Murtola
- School of Medicine, University of Tampere, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
50
|
Biancini GB, Morás AM, Reinhardt LS, Busatto FF, de Moura Sperotto ND, Saffi J, Moura DJ, Giugliani R, Vargas CR. Globotriaosylsphingosine induces oxidative DNA damage in cultured kidney cells. Nephrology (Carlton) 2017; 22:490-493. [DOI: 10.1111/nep.12977] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/16/2016] [Accepted: 11/30/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Giovana Brondani Biancini
- Graduate Program in Biological Sciences: Biochemistry; Federal University of Rio Grande do Sul (UFRGS); Porto Alegre RS Brazil
- Medical Genetics Service; Clinical Hospital of Porto Alegre (HCPA); Porto Alegre RS Brazil
| | - Ana Moira Morás
- Laboratory of Genetic Toxicology; Federal University of Life Sciences of Porto Alegre (UFSCPA); Porto Alegre RS Brazil
| | - Luiza Steffens Reinhardt
- Laboratory of Genetic Toxicology; Federal University of Life Sciences of Porto Alegre (UFSCPA); Porto Alegre RS Brazil
| | - Franciele Faccio Busatto
- Laboratory of Genetic Toxicology; Federal University of Life Sciences of Porto Alegre (UFSCPA); Porto Alegre RS Brazil
| | | | - Jenifer Saffi
- Laboratory of Genetic Toxicology; Federal University of Life Sciences of Porto Alegre (UFSCPA); Porto Alegre RS Brazil
| | - Dinara Jaqueline Moura
- Laboratory of Genetic Toxicology; Federal University of Life Sciences of Porto Alegre (UFSCPA); Porto Alegre RS Brazil
| | - Roberto Giugliani
- Graduate Program in Biological Sciences: Biochemistry; Federal University of Rio Grande do Sul (UFRGS); Porto Alegre RS Brazil
- Medical Genetics Service; Clinical Hospital of Porto Alegre (HCPA); Porto Alegre RS Brazil
- Department of Genetics; UFRGS; Porto Alegre RS Brazil
| | - Carmen Regla Vargas
- Graduate Program in Biological Sciences: Biochemistry; Federal University of Rio Grande do Sul (UFRGS); Porto Alegre RS Brazil
- Medical Genetics Service; Clinical Hospital of Porto Alegre (HCPA); Porto Alegre RS Brazil
- Graduate Program in Pharmaceutical Sciences; UFRGS; Porto Alegre RS Brazil
| |
Collapse
|