1
|
Viggiano D, Joshi R, Borriello G, Cacciola G, Gonnella A, Gigliotti A, Nigro M, Gigliotti G. SGLT2 Inhibitors: The First Endothelial-Protector for Diabetic Nephropathy. J Clin Med 2025; 14:1241. [PMID: 40004772 PMCID: PMC11856817 DOI: 10.3390/jcm14041241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Sodium-glucose co-transporter type 2 inhibitors (SGLT2i) have emerged as a class of agents relevant for managing diabetic nephropathy and cardiopathy. In a previous report, we noticed that these drugs share, with other drugs with "nephroprotective" effects, the ability to reduce the glomerular filtration rate (GFR), thus suggesting the kidney hemodynamic effect as a proxy for optimal drug dosage. We also noticed that all known nephroprotective drugs exert cardioprotective functions, suggesting the possibility of activities not mediated by the kidney. Finally, we observe that nephroprotective drugs can be grouped according to their effects on hemoglobin levels, thus suggesting their mechanism of action. While the primary mechanism of SGLT2i involves glycosuria and natriuria, growing evidence suggests broader therapeutic effects beyond hemodynamic modulation. Specifically, the evidence that SGLT2 can be expressed in several atypical regions under pathological conditions, supports the possibility that its inhibition has several extratubular effects. Evidence supports the hypothesis that SGLT2i influence mitochondrial function in various cell types affected by diabetes, particularly in the context of diabetic nephropathy. Notably, in SGLT2i-treated patients, the extent of albumin-creatinine ratio (ACR) reduction post-treatment may be correlated with mitochondrial staining intensity in glomerular endothelial cells. This implies that the anti-proteinuric effects of SGLT2i could involve direct actions on glomerular endothelial cell. Our investigation into the role of SGLT2 inhibitors (SGLT2i) in endothelial function suggests that the aberrant expression of SGLT2 in endothelial cells in T2DM would lead to intracellular accumulation of glucose; therefore, SGLT2i are the first type of endothelial protective drugs available today, with potential implications for ageing-related kidney disease. The review reveals two major novel findings: SGLT2 inhibitors are the first known class of endothelial-protective drugs, due to their ability to prevent glucose accumulation in endothelial cells where SGLT2 is aberrantly expressed in Type 2 Diabetes. Additionally, the research demonstrates that SGLT2 inhibitors share a GFR-reducing effect with other nephroprotective drugs, suggesting both a mechanism for optimal drug dosing and potential broader applications in ageing-related kidney disease through their effects on mitochondrial function and glomerular endothelial cells.
Collapse
Affiliation(s)
- Davide Viggiano
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Rashmi Joshi
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Gianmarco Borriello
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Giovanna Cacciola
- Department Translational Medical Sciences, University of Campania, 80138 Naples, Italy; (R.J.); (G.B.); (G.C.)
| | - Annalisa Gonnella
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| | - Andrea Gigliotti
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| | - Michelangelo Nigro
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| | - Giuseppe Gigliotti
- Department Nephrology, Eboli Hospital, 84025 Eboli, Italy; (A.G.); (A.G.); (M.N.); (G.G.)
| |
Collapse
|
2
|
Singh H, Singh R, Singh A, Singh H, Singh G, Kaur S, Singh B. Role of oxidative stress in diabetes-induced complications and their management with antioxidants. Arch Physiol Biochem 2024; 130:616-641. [PMID: 37571852 DOI: 10.1080/13813455.2023.2243651] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 08/13/2023]
Abstract
Diabetes mellitus (DM) is a huge global health issue and one of the most studied diseases, with a large global prevalence. Oxidative stress is a cytotoxic consequence of the excessive development of ROS and suppression of the antioxidant defense system for ROS elimination, which accelerates the progression of diabetes complications such as diabetic neuropathy, retinopathy, and nephropathy. Hyperglycaemia induced oxidative stress causes the activation of seven major pathways implicated in the pathogenesis of diabetic complications. These pathways increase the production of ROS and RNS, which contributes to dysregulated autophagy, gene expression changes, and the development of numerous pro-inflammatory mediators which may eventually lead to diabetic complications. This review will illustrate that oxidative stress plays a vital role in the pathogenesis of diabetic complications, and the use of antioxidants will help to reduce oxidative stress and thus may alleviate diabetic complications.
Collapse
Affiliation(s)
- Hasandeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Rajanpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Arshdeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Harshbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Gurpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Sarabjit Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| |
Collapse
|
3
|
Wang S, Song S, Gao J, Duo Y, Gao Y, Fu Y, Dong Y, Yuan T, Zhao W. Glycated haemoglobin variability and risk of renal function decline in type 2 diabetes mellitus: An updated systematic review and meta-analysis. Diabetes Obes Metab 2024; 26:5167-5182. [PMID: 39233504 DOI: 10.1111/dom.15861] [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: 06/20/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE To assess the association between glycated haemoglobin (HbA1c) variability and risk of renal function decline in type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS A comprehensive search was carried out in PubMed, Embase, Web of Science and the Cochrane Library (until 12 March 2024). The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement guidelines were followed for this meta-analysis. HbA1c variability was presented as indices of the standard deviation (SD), coefficient of variation (CV), HbA1c variability score (HVS) and haemoglobin glycation index (HGI). This meta-analysis was performed using random-effect models. RESULTS Eighteen studies met the objectives of this meta-analysis. The analyses showed positive associations between HbA1c variability and kidney function decline, with hazard ratio (HR) 1.26 (95% confidence interval [CI] 1.15-1.38) for high versus low SD groups, HR 1.47 (95% CI 1.30-1.65) for CV groups, HR 1.32 (95% CI 1.10-1.57) for HVS groups and HR 1.53 (95% CI 1.05-2.23) for HGI groups. In addition, each 1% increase in SD and CV was linked to kidney function decline, with HR 1.26 (95% CI 1.17-1.35), and 1.13 (95% CI 1.03-1.23), respectively. Also, each 1-SD increase in SD of HbA1c was associated with deterioration in renal function, with HR 1.17 (95% CI 1.07-1.29). CONCLUSIONS The four HbA1c variability indicators were all positively associated with renal function decline progression; therefore, HbA1c variability might play an important and promising role in guiding glycaemic control targets and predicting kidney function decline progression in T2DM.
Collapse
Affiliation(s)
- Shihan Wang
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuoning Song
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junxiang Gao
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanbei Duo
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuting Gao
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Fu
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingyue Dong
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weigang Zhao
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Ren L, Pushpakumar S, Almarshood H, Das SK, Sen U. Epigenetic DNA Methylation and Protein Homocysteinylation: Key Players in Hypertensive Renovascular Damage. Int J Mol Sci 2024; 25:11599. [PMID: 39519150 PMCID: PMC11546175 DOI: 10.3390/ijms252111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Hypertension has been a threat to the health of people, the mechanism of which, however, remains poorly understood. It is clinically related to loss of nephron function, glomerular sclerosis, or necrosis, resulting in renal functional declines. The mechanisms underlying hypertension's development and progression to organ damage, including hypertensive renal damage, remain to be fully elucidated. As a developing approach, epigenetics has been postulated to elucidate the phenomena that otherwise cannot be explained by genetic studies. The main epigenetic hallmarks, such as DNA methylation, histone acetylation, deacetylation, noncoding RNAs, and protein N-homocysteinylation have been linked with hypertension. In addition to contributing to endothelial dysfunction and oxidative stress, biologically active gases, including NO, CO, and H2S, are crucial regulators contributing to vascular remodeling since their complex interplay conducts homeostatic functions in the renovascular system. Importantly, epigenetic modifications also directly contribute to the pathogenesis of kidney damage via protein N-homocysteinylation. Hence, epigenetic modulation to intervene in renovascular damage is a potential therapeutic approach to treat renal disease and dysfunction. This review illustrates some of the epigenetic hallmarks and their mediators, which have the ability to diminish the injury triggered by hypertension and renal disease. In the end, we provide potential therapeutic possibilities to treat renovascular diseases in hypertension.
Collapse
Affiliation(s)
- Lu Ren
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Swapan K. Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| |
Collapse
|
5
|
Kushwaha K, Garg SS, Mandal D, Khurana N, Gupta J. Screening of natural epigenetic modifiers for managing glycemic memory and diabetic nephropathy. J Drug Target 2024; 32:807-819. [PMID: 38749010 DOI: 10.1080/1061186x.2024.2356737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/03/2024] [Accepted: 05/10/2024] [Indexed: 05/28/2024]
Abstract
Short hyperglycaemic episodes trigger metabolic memory (MM) in which managing hyperglycaemia alone is not enough to tackle the progression of Diabetic nephropathy on the epigenetic axis. We used a structural similarity search approach to identify phytochemicals similar to natural epigenetic modifiers and docked with SIRT1 protein and did ADME studies. We found that UMB was 84.3% similar to esculetin. Upon docking, we found that UMB had a binding energy of -9.2 kcal/mol while the standard ligand had -11.8 kcal/mol. ADME showed UMB to be a good lead. 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay showed it to be a good antioxidant with IC50 of 107 µg/mL and MTT stands for 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) showed that it does not promote cell death. Oxidative biomarkers in vitro showed UMB was able to ameliorate glycemic memory induced by high glucose. Western blot revealed decreased histone acetylation under hyperglycaemic conditions and upon treatment with UMB along with DR, its levels increased. This led us to check our hypothesis of whether concomitant diet reversal (DR) together with UMB can alleviate high-fat diet-induced metabolic memory and diabetic nephropathy (DN) in SD rats. UMB was able to decrease blood glucose, lipid, renal, and liver profile concluding UMB was able to ameliorate DN and MM by increasing the histone acetylation level.
Collapse
Affiliation(s)
- Kriti Kushwaha
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Sourbh Suren Garg
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Debojyoti Mandal
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Navneet Khurana
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
6
|
Yang Y, Meng L, Hu X, Li X. Renal functional outcomes after nephrectomy in patients with localized renal cell carcinoma and diabetes mellitus: a systematic review and meta-analysis. Int Urol Nephrol 2024; 56:1859-1868. [PMID: 38300449 DOI: 10.1007/s11255-023-03885-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/31/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION AND OBJECTIVE Diabetes mellitus (DM), one of the most common comorbidities in patients with renal cell carcinoma (RCC), was proven to be an important prognostic factor of overall survival for these patients. Regarding the influence on renal function after nephrectomy, evidence is still scant. This systematic review and meta-analysis was conducted to provide a more reliable analysis of the association between DM and long-term renal functional outcomes after nephrectomy. METHODS The PubMed, Web of Science, Embase and Cochrane Library (CENTRAL) databases were searched for eligible studies from inception to January 2023. Hazard ratios (HRs) with 95% confidence intervals (CIs) were extracted to evaluate the association between DM and renal functional outcomes using a random effects model. Stata 17.0 software was used for statistical analysis. RESULTS The meta-analysis included thirteen studies consisting of 8562 RCC patients who underwent nephrectomy. Preoperative comorbidity of DM was significantly associated with poor renal functional outcomes (HR = 1.91, 95% CI 1.48-2.48, p < 0.0001), regardless of ethnicity, follow-up time, body mass index (BMI) and age. However, in the radical nephrectomy subgroup, DM was not significantly associated with renal function decline (HR = 1.91, 95% CI 0.93-3.90, p = 0.0781). CONCLUSIONS The aggregate evidence indicated that preexisting DM may be associated with poor renal functional outcomes in patients with RCC after nephrectomy, especially in patients receiving partial nephrectomy. Urologists should focus more on the glycemic management of these patients after nephrectomy. More high-quality studies are needed to explore the influence of DM on renal function outcomes in postoperative patients.
Collapse
Affiliation(s)
- Yujia Yang
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Linghao Meng
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Xu Hu
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Li
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Yang T, Qi F, Guo F, Shao M, Song Y, Ren G, Linlin Z, Qin G, Zhao Y. An update on chronic complications of diabetes mellitus: from molecular mechanisms to therapeutic strategies with a focus on metabolic memory. Mol Med 2024; 30:71. [PMID: 38797859 PMCID: PMC11128119 DOI: 10.1186/s10020-024-00824-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Diabetes mellitus, a chronic metabolic disease, often leads to numerous chronic complications, significantly contributing to global morbidity and mortality rates. High glucose levels trigger epigenetic modifications linked to pathophysiological processes like inflammation, immunity, oxidative stress, mitochondrial dysfunction, senescence and various kinds of cell death. Despite glycemic control, transient hyperglycemia can persistently harm organs, tissues, and cells, a latent effect termed "metabolic memory" that contributes to chronic diabetic complications. Understanding metabolic memory's mechanisms could offer a new approach to mitigating these complications. However, key molecules and networks underlying metabolic memory remain incompletely understood. This review traces the history of metabolic memory research, highlights its key features, discusses recent molecules involved in its mechanisms, and summarizes confirmed and potential therapeutic compounds. Additionally, we outline in vitro and in vivo models of metabolic memory. We hope this work will inform future research on metabolic memory's regulatory mechanisms and facilitate the development of effective therapeutic compounds to prevent diabetic complications.
Collapse
Affiliation(s)
- Tongyue Yang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Feng Qi
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Research Institute of Nephrology, Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Feng Guo
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mingwei Shao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Song
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Gaofei Ren
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhao Linlin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Guijun Qin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yanyan Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
8
|
Litke R, Vicari J, Huang BT, Shapiro L, Roh KH, Silver A, Talreja P, Palacios N, Yoon Y, Kellner C, Kaniskan H, Vangeti S, Jin J, Ramos-Lopez I, Mobbs C. Novel small molecules inhibit proteotoxicity and inflammation: Mechanistic and therapeutic implications for Alzheimer's Disease, healthspan and lifespan- Aging as a consequence of glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544352. [PMID: 37398396 PMCID: PMC10312632 DOI: 10.1101/2023.06.12.544352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Inflammation drives many age-related, especially neurological, diseases, and likely mediates age-related proteotoxicity. For example, dementia due to Alzheimer's Disease (AD), cerebral vascular disease, many other neurodegenerative conditions is increasingly among the most devastating burdens on the American (and world) health system and threatens to bankrupt the American health system as the population ages unless effective treatments are developed. Dementia due to either AD or cerebral vascular disease, and plausibly many other neurodegenerative and even psychiatric conditions, is driven by increased age-related inflammation, which in turn appears to mediate Abeta and related proteotoxic processes. The functional significance of inflammation during aging is also supported by the fact that Humira, which is simply an antibody to the pro-inflammatory cytokine TNF-a, is the best-selling drug in the world by revenue. These observations led us to develop parallel high-throughput screens to discover small molecules which inhibit age-related Abeta proteotoxicity in a C. elegans model of AD AND LPS-induced microglial TNF-a. In the initial screen of 2560 compounds (Microsource Spectrum library) to delay Abeta proteotoxicity, the most protective compounds were, in order, phenylbutyrate, methicillin, and quetiapine, which belong to drug classes (HDAC inhibitors, beta lactam antibiotics, and tricyclic antipsychotics, respectably) already robustly implicated as promising to protect in neurodegenerative diseases, especially AD. RNAi and chemical screens indicated that the protective effects of HDAC inhibitors to reduce Abeta proteotoxicity are mediated by inhibition of HDAC2, also implicated in human AD, dependent on the HAT Creb binding protein (Cbp), which is also required for the protective effects of both dietary restriction and the daf-2 mutation (inactivation of IGF-1 signaling) during aging. In addition to methicillin, several other beta lactam antibiotics also delayed Abeta proteotoxicity and reduced microglial TNF-a. In addition to quetiapine, several other tricyclic antipsychotic drugs also delayed age-related Abeta proteotoxicity and increased microglial TNF-a, leading to the synthesis of a novel congener, GM310, which delays Abeta as well as Huntingtin proteotoxicity, inhibits LPS-induced mouse and human microglial and monocyte TNF-a, is highly concentrated in brain after oral delivery with no apparent toxicity, increases lifespan, and produces molecular responses highly similar to those produced by dietary restriction, including induction of Cbp inhibition of inhibitors of Cbp, and genes promoting a shift away from glycolysis and toward metabolism of alternate (e.g., lipid) substrates. GM310, as well as FDA-approved tricyclic congeners, prevented functional impairments and associated increase in TNF-a in a mouse model of stroke. Robust reduction of glycolysis by GM310 was functionally corroborated by flux analysis, and the glycolytic inhibitor 2-DG inhibited microglial TNF-a and other markers of inflammation, delayed Abeta proteotoxicity, and increased lifespan. These results support the value of phenotypic screens to discover drugs to treat age-related, especially neurological and even psychiatric diseases, including AD and stroke, and to clarify novel mechanisms driving neurodegeneration (e.g., increased microglial glycolysis drives neuroinflammation and subsequent neurotoxicity) suggesting novel treatments (selective inhibitors of microglial glycolysis).
Collapse
|
9
|
Liao Y, Chu C, Ma Q, Yan Y, Wang D, Sun Y, Wang Y, Hu J, Chen C, Mu J. Transient high salt intake causes epigenetic changes and leads to persistent inflammatory activation to produce "salt memory". J Nutr Biochem 2023; 115:109281. [PMID: 36758838 DOI: 10.1016/j.jnutbio.2023.109281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/21/2022] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Transient high salt intake causes a sustained increase in blood pressure (BP) even after returning to a normal-salt diet, a phenomenon known as "salt memory." However, the molecular mechanisms of this phenomenon remain to be elucidated. Dahl salt-sensitive (SS) rats were fed a high-salt (8% NaCl) or high-salt diet and treated with drugs for 8 to 16 weeks and then returned to a normal-salt diet for 3 months. This study investigated the molecular mechanisms of salt memory and its mediation of SS hypertension and renal damage. We show that transient high salt intake caused persistent elevation of BP and exacerbation of kidney damage in Dahl SS rats even after returning to a normal-salt diet. Both epigenetic changes and inflammatory activation also persisted after resumption of a normal diet. Arterial BP, renal injury and the inflammatory response returned to normal levels in rats administered mycophenolate mofetil (MMF) during the 8-week period of high salt intake, resulting in the disappearance of salt memory. However, the vasodilator hydralazine did not ameliorate kidney damage or inflammatory activation, although it decreased BP to control levels. Transient high salt intake increased histone 3 lysine 4 monomethylation (H3K4me1) levels at the nuclear factor κB (NF-κB) subunit p65 promoter in SS rats, promoting p65 gene transcription and NF-κB activation and further leading to a series of inflammatory responses. Our findings demonstrate that transient high salt-induced epigenetic changes and persistent inflammatory activation play important roles in salt memory and its mediation of SS hypertension and renal damage.
Collapse
Affiliation(s)
- Yueyuan Liao
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Chao Chu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Qiong Ma
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yu Yan
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Dan Wang
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yue Sun
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yang Wang
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Jiawen Hu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Chen Chen
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Jianjun Mu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China.
| |
Collapse
|
10
|
Knobel P, Litke R, Mobbs CV. Biological age and environmental risk factors for dementia and stroke: Molecular mechanisms. Front Aging Neurosci 2022; 14:1042488. [PMID: 36620763 PMCID: PMC9813958 DOI: 10.3389/fnagi.2022.1042488] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Since the development of antibiotics and vaccination, as well as major improvements in public hygiene, the main risk factors for morbidity and mortality are age and chronic exposure to environmental factors, both of which can interact with genetic predispositions. As the average age of the population increases, the prevalence and costs of chronic diseases, especially neurological conditions, are rapidly increasing. The deleterious effects of age and environmental risk factors, develop chronically over relatively long periods of time, in contrast to the relatively rapid deleterious effects of infectious diseases or accidents. Of particular interest is the hypothesis that the deleterious effects of environmental factors may be mediated by acceleration of biological age. This hypothesis is supported by evidence that dietary restriction, which universally delays age-related diseases, also ameliorates deleterious effects of environmental factors. Conversely, both age and environmental risk factors are associated with the accumulation of somatic mutations in mitotic cells and epigenetic modifications that are a measure of "biological age", a better predictor of age-related morbidity and mortality than chronological age. Here we review evidence that environmental risk factors such as smoking and air pollution may also drive neurological conditions, including Alzheimer's Disease, by the acceleration of biological age, mediated by cumulative and persistent epigenetic effects as well as somatic mutations. Elucidation of such mechanisms could plausibly allow the development of interventions which delay deleterious effects of both aging and environmental risk factors.
Collapse
Affiliation(s)
- Pablo Knobel
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rachel Litke
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charles V. Mobbs
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States,*Correspondence: Charles V. Mobbs,
| |
Collapse
|
11
|
Halladjian S, Kouril D, Miao H, Groller ME, Viola I, Isenberg T. Multiscale Unfolding: Illustratively Visualizing the Whole Genome at a Glance. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2022; 28:3456-3470. [PMID: 33705319 DOI: 10.1109/tvcg.2021.3065443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We present Multiscale Unfolding, an interactive technique for illustratively visualizing multiple hierarchical scales of DNA in a single view, showing the genome at different scales and demonstrating how one scale spatially folds into the next. The DNA's extremely long sequential structure-arranged differently on several distinct scale levels-is often lost in traditional 3D depictions, mainly due to its multiple levels of dense spatial packing and the resulting occlusion. Furthermore, interactive exploration of this complex structure is cumbersome, requiring visibility management like cut-aways. In contrast to existing temporally controlled multiscale data exploration, we allow viewers to always see and interact with any of the involved scales. For this purpose we separate the depiction into constant-scale and scale transition zones. Constant-scale zones maintain a single-scale representation, while still linearly unfolding the DNA. Inspired by illustration, scale transition zones connect adjacent constant-scale zones via level unfolding, scaling, and transparency. We thus represent the spatial structure of the whole DNA macro-molecule, maintain its local organizational characteristics, linearize its higher-level organization, and use spatially controlled, understandable interpolation between neighboring scales. We also contribute interaction techniques that provide viewers with a coarse-to-fine control for navigating within our all-scales-in-one-view representations and visual aids to illustrate the size differences. Overall, Multiscale Unfolding allows viewers to grasp the DNA's structural composition from chromosomes to the atoms, with increasing levels of "unfoldedness," and can be applied in data-driven illustration and communication.
Collapse
|
12
|
Khajah MA, Khushaish S, Luqmani YA. Glucose deprivation reduces proliferation and motility, and enhances the anti-proliferative effects of paclitaxel and doxorubicin in breast cell lines in vitro. PLoS One 2022; 17:e0272449. [PMID: 35917304 PMCID: PMC9345370 DOI: 10.1371/journal.pone.0272449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022] Open
Abstract
Background Breast cancer chemotherapy with high dose alkylating agents is severely limited by their collateral toxicity to crucial normal tissues such as immune and gut cells. Taking advantage of the selective dependence of cancer cells on high glucose and combining glucose deprivation with these agents could produce therapeutic synergy. Methods In this study we examined the effect of glucose as well as its deprivation, and antagonism using the non-metabolized analogue 2-deoxy glucose, on the proliferation of several breast cancer cell lines MCF7, MDA-MB-231, YS1.2 and pII and one normal breast cell line, using the MTT assay. Motility was quantitatively assessed using the wound healing assay. Lactate, as the end product of anaerobic glucose metabolism, secreted into culture medium was measured by a biochemical assay. The effect of paclitaxel and doxorubicin on cell proliferation was tested in the absence and presence of low concentrations of glucose using MTT assay. Results In all cell lines, glucose supplementation enhanced while glucose deprivation reduced both their proliferation and motility. Lactate added to the medium could substitute for glucose. The inhibitory effects of paclitaxel and doxorubicin were significantly enhanced when glucose concentration was decreased in the culture medium, requiring 1000-fold lesser concentration to achieve a similar degree of inhibition to that seen in glucose-containing medium. Conclusion Our data show that a synergy was obtained by combining paclitaxel and doxorubicin with glucose reduction to inhibit cancer cell growth, which in vivo, might be achieved by applying a carbohydrate-restricted diet during the limited phase of application of chemotherapy; this could permit a dose reduction of the cytotoxic agents, resulting in greater tolerance and lesser side effects.
Collapse
|
13
|
Liao Y, Chu C, Yan Y, Wang D, Ma Q, Gao K, Sun Y, Hu J, Zheng W, Mu J. High Dietary Salt Intake Is Associated With Histone Methylation in Salt-Sensitive Individuals. Front Nutr 2022; 9:857562. [PMID: 35571911 PMCID: PMC9097549 DOI: 10.3389/fnut.2022.857562] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background High salt diet is one of the important risk factors of hypertension and cardiovascular diseases. Increasingly strong evidence supports epigenetic mechanisms' significant role in hypertension. We aimed to explore associations of epigenetics with high salt diet, salt sensitivity (SS), and SS hypertension. Methods We conducted a dietary intervention study of chronic salt loading in 339 subjects from northern China in 2004 and divided the subjects into different salt sensitivity phenotypes. A total of 152 participants were randomly selected from the same cohort for follow-up in 2018 to explore the effect of a high-salt diet on serum monomethylation of H3K4 (H3K4me1), histone methyltransferase Set7, and lysine-specific demethylase 1 (LSD-1). Results Among SS individuals, the blood pressure (SBP: 140.8 vs. 132.9 mmHg; MAP: 104.2 vs. 98.7 mmHg) and prevalence of hypertension (58.8 vs. 32.8%) were significantly higher in high salt (HS) diet group than in normal salt (NS) diet group, but not in the salt-resistant (SR) individuals (P > 0.05). Serum H3K4me1 level (287.3 vs. 179.7 pg/ml, P < 0.05) significantly increased in HS group of SS individuals, but not in SR individuals. We found daily salt intake in SS individuals was positively correlated with serum H3K4me1 (r = 0.322, P = 0.005) and Set7 (r = 0.340, P = 0.005) levels after adjusting for age and gender, but not with LSD-1 (r = -0.137, P = 0.251). In addition, positive correlation between the serum H3K4me1 level and Set7 level (r = 0.326, P = 0.007) was also found in SS individuals. These correlations were not evident in SR individuals. Conclusion Our study indicates that high salt diet increases the serum H3K4me1 and Set7 levels in SS individuals.
Collapse
Affiliation(s)
- Yueyuan Liao
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Chao Chu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Yu Yan
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Dan Wang
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Qiong Ma
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Ke Gao
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Yue Sun
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Jiawen Hu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Wenling Zheng
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Jianjun Mu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| |
Collapse
|
14
|
A Novel Glucose-Sensitive Scaffold Accelerates Osteogenesis in Diabetic Conditions. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4133562. [PMID: 35342759 PMCID: PMC8956406 DOI: 10.1155/2022/4133562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 02/10/2022] [Accepted: 03/06/2022] [Indexed: 02/05/2023]
Abstract
Mandibular bone regeneration is still a big challenge in those diabetic patients with poorly controlled blood glucose. In this study, we prepared a novel glucose-sensitive controlled-release fiber scaffold (PVA-HTCC/PEO-rhBMP2-glucose oxidase (PHPB-G)), which contained the recombinant human bone morphogenetic protein 2 (rhBMP2) by coaxial cospinning and grafted with glucose oxidase (GOD). We presented evidence that PHPB-G could undergo a series of structural changes with the blood glucose and promoted bone regeneration in diabetic rat. PHPB-G expanded the voids in nanofibers when blood glucose levels elevated. More importantly, its slow-release rhBMP2 effectively promoted the healing of bone defects. These data suggested that the PHPB-G delivery system may provide a potential treatment strategy for patients with severe diabetic alveolar bone defects.
Collapse
|
15
|
Gao P, You M, Li L, Zhang Q, Fang X, Wei X, Zhou Q, Zhang H, Wang M, Lu Z, Wang L, Sun F, Liu D, Zheng H, Yan Z, Yang G, Zhu Z. Salt-Induced Hepatic Inflammatory Memory Contributes to Cardiovascular Damage Through Epigenetic Modulation of SIRT3. Circulation 2022; 145:375-391. [PMID: 35100024 DOI: 10.1161/circulationaha.121.055600] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND High salt intake is the leading dietary risk factor for cardiovascular diseases. Although clinical evidence suggests that high salt intake is associated with nonalcoholic fatty liver disease, which is an independent risk factor for cardiovascular diseases, it remains elusive whether salt-induced hepatic damage leads to the development of cardiovascular diseases. METHODS Mice were fed with normal or high-salt diet for 8 weeks to determine the effect of salt loading on liver histological changes and blood pressure, and salt withdrawal and metformin treatment were also conducted on some high-salt diet-fed mice. Adeno-associated virus 8, global knockout, or tissue-specific knockout mice were used to manipulate the expression of some target genes in vivo, including SIRT3 (sirtuin 3), NRF2 (NF-E2-related factor 2), and AMPK (AMP-activated protein kinase). RESULTS Mice fed with a high-salt diet displayed obvious hepatic steatosis and inflammation, accompanied with hypertension and cardiac dysfunction. All these pathological changes persisted after salt withdrawal, displaying a memory phenomenon. Gene expression analysis and phenotypes of SIRT3 knockout mice revealed that reduced expression of SIRT3 was a chief culprit responsible for the persistent inflammation in the liver, and recovering SIRT3 expression in the liver effectively inhibits the sustained hepatic inflammation and cardiovascular damage. Mechanistical studies reveal that high salt increases acetylated histone 3 lysine 27 (H3K27ac) on SIRT3 promoter in hepatocytes, thus inhibiting the binding of NRF2, and results in the sustained inhibition of SIRT3 expression. Treatment with metformin activated AMPK, which inhibited salt-induced hepatic inflammatory memory and cardiovascular damage by lowering the H3K27ac level on SIRT3 promoter, and increased NRF2 binding ability to activate SIRT3 expression. CONCLUSIONS This study demonstrates that SIRT3 inhibition caused by histone modification is the key factor for the persistent hepatic steatosis and inflammation that contributes to cardiovascular damage under high salt loading. Avoidance of excessive salt intake and active intervention of epigenetic modification may help to stave off the persistent inflammatory status that underlies high-salt-induced cardiovascular damage in clinical practice.
Collapse
Affiliation(s)
- Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Mei You
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Qin Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, China (Q. Zhang, X.F., M.W., G.Y.)
| | - Xia Fang
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital (H.Z.), Army Medical University, Chongqing China
| | - Xiao Wei
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Qing Zhou
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Hexuan Zhang
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital (H.Z.), Army Medical University, Chongqing China
| | - Miao Wang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, China (Q. Zhang, X.F., M.W., G.Y.)
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Lijuan Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Hongting Zheng
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, China (Q. Zhang, X.F., M.W., G.Y.)
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension (P.G., M.Y., L.L., X.W., Q. Zhou, H.Z., Z.L., L.W., F.S., D.L., Z.Y., Z.Z.), Army Medical University, Chongqing China
| |
Collapse
|
16
|
Sheng X, Guan Y, Ma Z, Wu J, Liu H, Qiu C, Vitale S, Miao Z, Seasock MJ, Palmer M, Shin MK, Duffin KL, Pullen SS, Edwards TL, Hellwege JN, Hung AM, Li M, Voight BF, Coffman TM, Brown CD, Susztak K. Mapping the genetic architecture of human traits to cell types in the kidney identifies mechanisms of disease and potential treatments. Nat Genet 2021; 53:1322-1333. [PMID: 34385711 PMCID: PMC9338440 DOI: 10.1038/s41588-021-00909-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
The functional interpretation of genome-wide association studies (GWAS) is challenging due to the cell-type-dependent influences of genetic variants. Here, we generated comprehensive maps of expression quantitative trait loci (eQTLs) for 659 microdissected human kidney samples and identified cell-type-eQTLs by mapping interactions between cell type abundances and genotypes. By partitioning heritability using stratified linkage disequilibrium score regression to integrate GWAS with single-cell RNA sequencing and single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing data, we prioritized proximal tubules for kidney function and endothelial cells and distal tubule segments for blood pressure pathogenesis. Bayesian colocalization analysis nominated more than 200 genes for kidney function and hypertension. Our study clarifies the mechanism of commonly used antihypertensive and renal-protective drugs and identifies drug repurposing opportunities for kidney disease.
Collapse
Affiliation(s)
- Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuting Guan
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziyuan Ma
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junnan Wu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongbo Liu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Vitale
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhen Miao
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew J Seasock
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Kevin L Duffin
- Eli Lilly and Company Lilly Corporate Center, Indianapolis, IN, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jacklyn N Hellwege
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adriana M Hung
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mingyao Li
- Department of Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Benjamin F Voight
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas M Coffman
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | | | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA.
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Tang G, Li S, Zhang C, Chen H, Wang N, Feng Y. Clinical efficacies, underlying mechanisms and molecular targets of Chinese medicines for diabetic nephropathy treatment and management. Acta Pharm Sin B 2021; 11:2749-2767. [PMID: 34589395 PMCID: PMC8463270 DOI: 10.1016/j.apsb.2020.12.020] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/17/2020] [Accepted: 12/25/2020] [Indexed: 12/17/2022] Open
Abstract
Diabetic nephropathy (DN) has been recognized as a severe complication of diabetes mellitus and a dominant pathogeny of end-stage kidney disease, which causes serious health problems and great financial burden to human society worldwide. Conventional strategies, such as renin-angiotensin-aldosterone system blockade, blood glucose level control, and bodyweight reduction, may not achieve satisfactory outcomes in many clinical practices for DN management. Notably, due to the multi-target function, Chinese medicine possesses promising clinical benefits as primary or alternative therapies for DN treatment. Increasing studies have emphasized identifying bioactive compounds and molecular mechanisms of reno-protective effects of Chinese medicines. Signaling pathways involved in glucose/lipid metabolism regulation, antioxidation, anti-inflammation, anti-fibrosis, and podocyte protection have been identified as crucial mechanisms of action. Herein, we summarize the clinical efficacies of Chinese medicines and their bioactive components in treating and managing DN after reviewing the results demonstrated in clinical trials, systematic reviews, and meta-analyses, with a thorough discussion on the relative underlying mechanisms and molecular targets reported in animal and cellular experiments. We aim to provide comprehensive insights into the protective effects of Chinese medicines against DN.
Collapse
Key Words
- ACEI, angiotensin-converting enzyme inhibitor
- ADE, adverse event
- AGEs, advanced glycation end-products
- AM, mesangial area
- AMPKα, adenosine monophosphate-activated protein kinase α
- ARB, angiotensin receptor blocker
- AREs, antioxidant response elements
- ATK, protein kinase B
- BAX, BCL-2-associated X protein
- BCL-2, B-cell lymphoma 2
- BCL-XL, B-cell lymphoma-extra large
- BMP-7, bone morphogenetic protein-7
- BUN, blood urea nitrogen
- BW, body weight
- C, control group
- CCR, creatinine clearance rate
- CD2AP, CD2-associated protein
- CHOP, C/EBP homologous protein
- CI, confidence interval
- COL-I/IV, collagen I/IV
- CRP, C-reactive protein
- CTGF, connective tissue growth factor
- Chinese medicine
- D, duration
- DAG, diacylglycerol
- DG, glomerular diameter
- DKD, diabetic kidney disease
- DM, diabetes mellitus
- DN, diabetic nephropathy
- Diabetic kidney disease
- Diabetic nephropathy
- EMT, epithelial-to-mesenchymal transition
- EP, E-prostanoid receptor
- ER, endoplasmic reticulum
- ESRD, end-stage renal disease
- ET-1, endothelin-1
- ETAR, endothelium A receptor
- FBG, fasting blood glucose
- FN, fibronectin
- GCK, glucokinase
- GCLC, glutamate-cysteine ligase catalytic subunit
- GFR, glomerular filtration rate
- GLUT4, glucose transporter type 4
- GPX, glutathione peroxidase
- GRB 10, growth factor receptor-bound protein 10
- GRP78, glucose-regulated protein 78
- GSK-3, glycogen synthase kinase 3
- Gαq, Gq protein alpha subunit
- HDL-C, high density lipoprotein-cholesterol
- HO-1, heme oxygenase-1
- HbA1c, glycosylated hemoglobin
- Herbal medicine
- ICAM-1, intercellular adhesion molecule-1
- IGF-1, insulin-like growth factor 1
- IGF-1R, insulin-like growth factor 1 receptor
- IKK-β, IκB kinase β
- IL-1β/6, interleukin 1β/6
- IR, insulin receptor
- IRE-1α, inositol-requiring enzyme-1α
- IRS, insulin receptor substrate
- IκB-α, inhibitory protein α
- JAK, Janus kinase
- JNK, c-Jun N-terminal kinase
- LC3, microtubule-associated protein light chain 3
- LDL, low-density lipoprotein
- LDL-C, low density lipoprotein-cholesterol
- LOX1, lectin-like oxidized LDL receptor 1
- MAPK, mitogen-activated protein kinase
- MCP-1, monocyte chemotactic protein-1
- MD, mean difference
- MDA, malondialdehyde
- MMP-2, matrix metallopeptidase 2
- MYD88, myeloid differentiation primary response 88
- Molecular target
- N/A, not applicable
- N/O, not observed
- N/R, not reported
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NOX-4, nicotinamide adenine dinucleotide phosphate-oxidase-4
- NQO1, NAD(P)H:quinone oxidoreductase 1
- NRF2, nuclear factor erythroid 2-related factor 2
- OCP, oxidative carbonyl protein
- ORP150, 150-kDa oxygen-regulated protein
- P70S6K, 70-kDa ribosomal protein S6 kinase
- PAI-1, plasminogen activator inhibitor-1
- PARP, poly(ADP-Ribose) polymerase
- PBG, postprandial blood glucose
- PERK, protein kinase RNA-like eukaryotic initiation factor 2A kinase
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1α
- PGE2, prostaglandin E2
- PI3K, phosphatidylinositol 3 kinases
- PINK1, PTEN-induced putative kinase 1
- PKC, protein kinase C
- PTEN, phosphatase and tensin homolog
- RAGE, receptors of AGE
- RASI, renin-angiotensin system inhibitor
- RCT, randomized clinical trial
- ROS, reactive oxygen species
- SCr, serum creatinine
- SD, standard deviation
- SD-rat, Sprague–Dawley rat
- SIRT1, sirtuin 1
- SMAD, small mothers against decapentaplegic
- SMD, standard mean difference
- SMURF-2, SMAD ubiquitination regulatory factor 2
- SOCS, suppressor of cytokine signaling proteins
- SOD, superoxide dismutase
- STAT, signal transducers and activators of transcription
- STZ, streptozotocin
- Signaling pathway
- T, treatment group
- TBARS, thiobarbituric acid-reactive substance
- TC, total cholesterol
- TCM, traditional Chinese medicine
- TFEB, transcription factor EB
- TG, triglyceride
- TGBM, thickness of glomerular basement membrane
- TGF-β, tumor growth factor β
- TGFβR-I/II, TGF-β receptor I/II
- TII, tubulointerstitial injury index
- TLR-2/4, toll-like receptor 2/4
- TNF-α, tumor necrosis factor α
- TRAF5, tumor-necrosis factor receptor-associated factor 5
- UACR, urinary albumin to creatinine ratio
- UAER, urinary albumin excretion rate
- UMA, urinary microalbumin
- UP, urinary protein
- VCAM-1, vascular cell adhesion molecule-1
- VEGF, vascular endothelial growth factor
- WMD, weight mean difference
- XBP-1, spliced X box-binding protein 1
- cAMP, cyclic adenosine monophosphate
- eGFR, estimated GFR
- eIF2α, eukaryotic initiation factor 2α
- mTOR, mammalian target of rapamycin
- p-IRS1, phospho-IRS1
- p62, sequestosome 1 protein
- α-SMA, α smooth muscle actin
Collapse
Affiliation(s)
- Guoyi Tang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Haiyong Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
18
|
Demir S, Nawroth PP, Herzig S, Ekim Üstünel B. Emerging Targets in Type 2 Diabetes and Diabetic Complications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100275. [PMID: 34319011 PMCID: PMC8456215 DOI: 10.1002/advs.202100275] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/07/2021] [Indexed: 05/06/2023]
Abstract
Type 2 diabetes is a metabolic, chronic disorder characterized by insulin resistance and elevated blood glucose levels. Although a large drug portfolio exists to keep the blood glucose levels under control, these medications are not without side effects. More importantly, once diagnosed diabetes is rarely reversible. Dysfunctions in the kidney, retina, cardiovascular system, neurons, and liver represent the common complications of diabetes, which again lack effective therapies that can reverse organ injury. Overall, the molecular mechanisms of how type 2 diabetes develops and leads to irreparable organ damage remain elusive. This review particularly focuses on novel targets that may play role in pathogenesis of type 2 diabetes. Further research on these targets may eventually pave the way to novel therapies for the treatment-or even the prevention-of type 2 diabetes along with its complications.
Collapse
Affiliation(s)
- Sevgican Demir
- Institute for Diabetes and Cancer (IDC)Helmholtz Center MunichIngolstädter Landstr. 1Neuherberg85764Germany
- Joint Heidelberg ‐ IDC Translational Diabetes ProgramInternal Medicine 1Heidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
- DZDDeutsches Zentrum für DiabetesforschungIngolstädter Landstraße 1Neuherberg85764Germany
- Department of Internal Medicine 1 and Clinical ChemistryHeidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
| | - Peter P. Nawroth
- Institute for Diabetes and Cancer (IDC)Helmholtz Center MunichIngolstädter Landstr. 1Neuherberg85764Germany
- Joint Heidelberg ‐ IDC Translational Diabetes ProgramInternal Medicine 1Heidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
- DZDDeutsches Zentrum für DiabetesforschungIngolstädter Landstraße 1Neuherberg85764Germany
- Department of Internal Medicine 1 and Clinical ChemistryHeidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC)Helmholtz Center MunichIngolstädter Landstr. 1Neuherberg85764Germany
- Joint Heidelberg ‐ IDC Translational Diabetes ProgramInternal Medicine 1Heidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
- DZDDeutsches Zentrum für DiabetesforschungIngolstädter Landstraße 1Neuherberg85764Germany
- Department of Internal Medicine 1 and Clinical ChemistryHeidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
| | - Bilgen Ekim Üstünel
- Institute for Diabetes and Cancer (IDC)Helmholtz Center MunichIngolstädter Landstr. 1Neuherberg85764Germany
- Joint Heidelberg ‐ IDC Translational Diabetes ProgramInternal Medicine 1Heidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
- DZDDeutsches Zentrum für DiabetesforschungIngolstädter Landstraße 1Neuherberg85764Germany
- Department of Internal Medicine 1 and Clinical ChemistryHeidelberg University HospitalIm Neuenheimer Feld 410Heidelberg69120Germany
| |
Collapse
|
19
|
Changes in metabolic syndrome status affect the incidence of end-stage renal disease in the general population: a nationwide cohort study. Sci Rep 2021; 11:1957. [PMID: 33479302 PMCID: PMC7820283 DOI: 10.1038/s41598-021-81396-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 12/16/2020] [Indexed: 01/21/2023] Open
Abstract
Few studies have investigated the impact of a change in metabolic syndrome (MetS) components on clinical renal outcomes in the general population. Using nationally representative data from the Korean National Health Insurance System, 13,310,924 subjects who underwent two health examinations over 2 years and were free from end-stage renal disease (ESRD) from 2009 to 2012 were followed to the end of 2016. The subjects were divided into four groups according to the change in MetS components between the two visits over 2 years: no MetS (–/–), post-MetS (–/+), pre-MetS (+/–), and both MetS (+/+). After a median follow up of 5.11 years, 18,582 incident ESRD cases were identified. In the multivariate adjusted model, the hazard ratio (HR) and 95% confidence interval (CI) for the development of ESRD in the both-MetS (+/+) group compared with the no-MetS (–/–) group was 5.65 (95% CI, 5.42–5.89), which was independent of age, sex, and baseline estimated glomerular filtration rate. Additionally, the HR for the pre-MetS (+/–) group versus the no-MetS (–/–) group was 2.28 (2.15–2.42). In subgroup analysis according to renal function, the impact of a change in MetS on the incidence of ESRD was more pronounced in individuals with advanced renal dysfunction. Subjects with resolved MetS components had a decreased risk of ESRD, but not as low as those that never had MetS components. This provides evidence supporting the strategy of modulating MetS in the general population to prevent the development of ESRD.
Collapse
|
20
|
Scisciola L, Rizzo MR, Marfella R, Cataldo V, Fontanella RA, Boccalone E, Paolisso G, Barbieri M. New insight in molecular mechanisms regulating SIRT6 expression in diabetes: Hyperglycaemia effects on SIRT6 DNA methylation. J Cell Physiol 2020; 236:4604-4613. [PMID: 33251641 DOI: 10.1002/jcp.30185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
Conflicting data are reported on the relationship between hyperglycaemia, diabetes and SIRT6 expression. To elucidate hyperglycaemia-induced molecular mechanisms regulating SIRT6 expression, the effect of hyperglycaemia on DNA methylation and SIRT6 expression has been evaluated in human aortic endothelial cells exposed to high glucose. DNA methylation of SIRT6 and any potential clinical implication was also evaluated in type 2 diabetic patients and compared with healthy controls. Endothelial cells exposed to high glucose showed lower methylation levels in SIRT6 promoter and increased SIRT6 and TET2 expression. The high glucose-induced epigenetic changes persisted after 48 h of glucose normalization. Diabetic patients showed lower levels of SIRT6 DNA methylation compared with nondiabetic patients. SIRT6 DNA methylation levels inversely correlated with plasma glucose. Our results firstly demonstrate the involvement of epigenetic mechanisms in regulating SIRT6 expression. Further experiments are necessary to clarify metabolic memory mechanisms driving to diabetic complications and how SIRT6 is potentially involved.
Collapse
Affiliation(s)
- Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Maria R Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Vittoria Cataldo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Rosaria A Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Eugenio Boccalone
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", L. Vanvitelli, Naples, Italy
| |
Collapse
|
21
|
Epigenetic Modifiers as Potential Therapeutic Targets in Diabetic Kidney Disease. Int J Mol Sci 2020; 21:ijms21114113. [PMID: 32526941 PMCID: PMC7312774 DOI: 10.3390/ijms21114113] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease is one of the fastest growing causes of death worldwide. Epigenetic regulators control gene expression and are potential therapeutic targets. There is functional interventional evidence for a role of DNA methylation and the histone post-translational modifications-histone methylation, acetylation and crotonylation-in the pathogenesis of kidney disease, including diabetic kidney disease. Readers of epigenetic marks, such as bromodomain and extra terminal (BET) proteins, are also therapeutic targets. Thus, the BD2 selective BET inhibitor apabetalone was the first epigenetic regulator to undergo phase-3 clinical trials in diabetic kidney disease with an endpoint of kidney function. The direct therapeutic modulation of epigenetic features is possible through pharmacological modulators of the specific enzymes involved and through the therapeutic use of the required substrates. Of further interest is the characterization of potential indirect effects of nephroprotective drugs on epigenetic regulation. Thus, SGLT2 inhibitors increase the circulating and tissue levels of β-hydroxybutyrate, a molecule that generates a specific histone modification, β-hydroxybutyrylation, which has been associated with the beneficial health effects of fasting. To what extent this impact on epigenetic regulation may underlie or contribute to the so-far unclear molecular mechanisms of cardio- and nephroprotection offered by SGLT2 inhibitors merits further in-depth studies.
Collapse
|
22
|
Mossel DM, Moganti K, Riabov V, Weiss C, Kopf S, Cordero J, Dobreva G, Rots MG, Klüter H, Harmsen MC, Kzhyshkowska J. Epigenetic Regulation of S100A9 and S100A12 Expression in Monocyte-Macrophage System in Hyperglycemic Conditions. Front Immunol 2020; 11:1071. [PMID: 32582175 PMCID: PMC7280556 DOI: 10.3389/fimmu.2020.01071] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
The number of diabetic patients in Europe and world-wide is growing. Diabetes confers a 2-fold higher risk for vascular disease. Lack of insulin production (Type 1 diabetes, T1D) or lack of insulin responsiveness (Type 2 diabetes, T2D) causes systemic metabolic changes such as hyperglycemia (HG) which contribute to the pathology of diabetes. Monocytes and macrophages are key innate immune cells that control inflammatory reactions associated with diabetic vascular complications. Inflammatory programming of macrophages is regulated and maintained by epigenetic mechanisms, in particular histone modifications. The aim of our study was to identify the epigenetic mechanisms involved in the hyperglycemia-mediated macrophage activation. Using Affymetrix microarray profiling and RT-qPCR we identified that hyperglycemia increased the expression of S100A9 and S100A12 in primary human macrophages. Expression of S100A12 was sustained after glucose levels were normalized. Glucose augmented the response of macrophages to Toll-like receptor (TLR)-ligands Palmatic acid (PA) and Lipopolysaccharide (LPS) i.e., pro-inflammatory stimulation. The abundance of activating histone Histone 3 Lysine 4 methylation marks (H3K4me1, H3K4me3) and general acetylation on histone 3 (AceH3) with the promoters of these genes was analyzed by chromatin immunoprecipitation. Hyperglycemia increased acetylation of histones bound to the promoters of S100A9 and S100A12 in M1 macrophages. In contrast, hyperglycemia caused a reduction in total H3 which correlated with the increased expression of both S100 genes. The inhibition of histone methyltransferases SET domain-containing protein (SET)7/9 and SET and MYND domain-containing protein (SMYD)3 showed that these specifically regulated S100A12 expression. We conclude that hyperglycemia upregulates expression of S100A9, S100A12 via epigenetic regulation and induces an activating histone code on the respective gene promoters in M1 macrophages. Mechanistically, this regulation relies on action of histone methyltransferases SMYD3 and SET7/9. The results define an important role for epigenetic regulation in macrophage mediated inflammation in diabetic conditions.
Collapse
Affiliation(s)
- Dieuwertje M Mossel
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Kondaiah Moganti
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany.,Department of Dermatology, University of Münster, Münster, Germany
| | - Vladimir Riabov
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics, Biomathematics and Information Processing, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Kopf
- Department of Medicine I: Endocrinology and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Julio Cordero
- Anatomy and Developmental Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Anatomy and Developmental Biology, CBTM, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marianne G Rots
- Department Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Harald Klüter
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Martin C Harmsen
- Department Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Julia Kzhyshkowska
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany.,German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| |
Collapse
|
23
|
Al-Hasani K, Khurana I, Farhat T, Eid A, El-Osta A. Epigenetics of Diabetic Nephropathy: From Biology to Therapeutics. EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/19-00137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Diabetic nephropathy (DN) is a lethal microvascular complication associated with Type 1 and Type 2 diabetes mellitus, and is the leading single cause of end-stage renal disease. Although genetic influences are important, epigenetic mechanisms have been implicated in several aspects of the disease. The current therapeutic methods to treat DN are limited to slowing disease progression without repair and regeneration of the damaged nephrons. Replacing dying or diseased kidney cells with new nephrons is an attractive strategy. This review considers the genetic and epigenetic control of nephrogenesis, together with the epigenetic mechanisms that accompany kidney development and recent advances in induced reprogramming and kidney cell regeneration in the context of DN.
Collapse
Affiliation(s)
- Keith Al-Hasani
- Department of Diabetes, Epigenetics in Human Health and Disease Laboratory, Monash University, Melbourne, Australia
| | - Ishant Khurana
- Department of Diabetes, Epigenetics in Human Health and Disease Laboratory, Monash University, Melbourne, Australia
| | - Theresa Farhat
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assam El-Osta
- Department of Diabetes, Epigenetics in Human Health and Disease Laboratory, Monash University, Melbourne, Australia; Department of Clinical Pathology, The University of Melbourne, Victoria, Australia; Faculty of Health, Department of Technology, Biomedical Laboratory Science, University College Copenhagen, Copenhagen, Denmark; Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong
| |
Collapse
|
24
|
Lee CL, Chen CH, Wu MJ, Tsai SF. The variability of glycated hemoglobin is associated with renal function decline in patients with type 2 diabetes. Ther Adv Chronic Dis 2020; 11:2040622319898370. [PMID: 32166009 PMCID: PMC7054736 DOI: 10.1177/2040622319898370] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Background The effect of glucose control, especially variability of glycated hemoglobin (HbA1c), on estimated glomerular filtration rate (eGFR) decline in type 2 diabetes is still debatable. Methods We used tertiles of coefficient of variation (CV) to determine the variability of HbA1c (HbA1c_CV). Mixed model repeated measures (MMRM) were used to evaluate the annual eGFR decline rate. Results In 1383 type 2 diabetic patients, we found the greater the HbA1c_CV, the greater the eGFR decline (p = 0.01, -0.99 in low, -1.73 in mid, and -2.53 ml/min/1.73 m2/year in high HbA1c_CV). Regardless of eGFR (⩾60 or <60 ml/min/1.73 m2), the same result holds (p = 0.019 and p = 0.007, respectively). In subgroup analysis of baseline HbA1c (%) (HbA1c < 7, 7 ⩽ HbA1c < 9, and HbA1c ⩾ 9), tertiles of HbA1c_CV showed similar effects on annual decline of eGFR (p = 0.193, 0.300, 0.182, respectively), although a trend for a steeper decline in renal function in the highest HbA1c_CV tertile was observed for all HbA1c strata, and even for HbA1c < 7%. A similar behavior was observed in patients with macroalbuminuria or normoalbuminuria (p = 0.219, and 0.109, respectively), with a significant trend in those with microalbuminuria (p = 0.019). Even in patients with HbA1c < 7, high HbA1c_CV also predicts rapid eGFR decline. Before macroalbuminuria, minimizing HbA1c_CV also has renal benefit. Conclusions HbA1c variability is an independent risk factor for deterioration of renal function. Even with well-controlled HbA1c levels (<7%), patients with high HbA1c_CV still experienced faster eGFR decline. Early minimization of glycemic variability (before macroalbuminuira) can curb deterioration of renal function. Monitoring and lowering of HbA1c_CV is highly recommended for diabetic care.
Collapse
Affiliation(s)
- Chia-Lin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| | - Cheng-Hsu Chen
- Department of Life Science, Tunghai University, Taichung
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taiwan Taichung Veterans General Hospital, Taichung
| | - Shang-Feng Tsai
- Division of Nephrology, Department of Internal Medicine, Taiwan Taichung Veterans General Hospital, No.160, Sec. 3, Taiwan Boulevard, Taichung 407
| |
Collapse
|
25
|
Kato M, Natarajan R. Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat Rev Nephrol 2020; 15:327-345. [PMID: 30894700 DOI: 10.1038/s41581-019-0135-6] [Citation(s) in RCA: 368] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The development and progression of diabetic kidney disease (DKD), a highly prevalent complication of diabetes mellitus, are influenced by both genetic and environmental factors. DKD is an important contributor to the morbidity of patients with diabetes mellitus, indicating a clear need for an improved understanding of disease aetiology to inform the development of more efficacious treatments. DKD is characterized by an accumulation of extracellular matrix, hypertrophy and fibrosis in kidney glomerular and tubular cells. Increasing evidence shows that genes associated with these features of DKD are regulated not only by classical signalling pathways but also by epigenetic mechanisms involving chromatin histone modifications, DNA methylation and non-coding RNAs. These mechanisms can respond to changes in the environment and, importantly, might mediate the persistent long-term expression of DKD-related genes and phenotypes induced by prior glycaemic exposure despite subsequent glycaemic control, a phenomenon called metabolic memory. Detection of epigenetic events during the early stages of DKD could be valuable for timely diagnosis and prompt treatment to prevent progression to end-stage renal disease. Identification of epigenetic signatures of DKD via epigenome-wide association studies might also inform precision medicine approaches. Here, we highlight the emerging role of epigenetics and epigenomics in DKD and the translational potential of candidate epigenetic factors and non-coding RNAs as biomarkers and drug targets for DKD.
Collapse
Affiliation(s)
- Mitsuo Kato
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
26
|
Abstract
Background Organisms can be primed by metabolic exposures to continue expressing response genes even once the metabolite is no longer available, and can affect the speed and magnitude of responsive gene expression during subsequent exposures. This “metabolic transcriptional memory” can have a profound impact on the survivability of organisms in fluctuating environments. Scope of review Here I present several examples of metabolic transcriptional memory in the microbial world and discuss what is known so far regarding the underlying mechanisms, which mainly focus on chromatin modifications, protein inheritance, and broad changes in metabolic network. From these lessons learned in microbes, some insights into the yet understudied human metabolic memory can be gained. I thus discuss the implications of metabolic memory in disease progression in humans – i.e., the memory of high blood sugar exposure and the resulting effects on diabetic complications. Major conclusions Carbon source shifts from glucose to other less preferred sugars such as lactose, galactose, and maltose for energy metabolism as well as starvation of a signal transduction precursor sugar inositol are well-studied examples of metabolic transcriptional memory in Escherichia coli and Saccharomyces cerevisiae. Although the specific factors guiding metabolic transcriptional memory are not necessarily conserved from microbes to humans, the same basic mechanisms are in play, as is observed in hyperglycemic memory. Exploration of new metabolic transcriptional memory systems as well as further detailed mechanistic analyses of known memory contexts in microbes is therefore central to understanding metabolic memory in humans, and may be of relevance for the successful treatment of the ever-growing epidemic of diabetes. Metabolic exposures can prime genes to have memory. Memory of carbon source shifts occurs in all kingdoms of life. Memory is maintained through multiple mechanisms including chromatin modifications, proteins, and metabolic network. Metabolic transcriptional memory in unicellular organisms is a part of “bet-hedging” strategies to ensure survival. Hyperglycemic memory in humans contributes to diabetes and aging.
Collapse
Affiliation(s)
- Poonam Bheda
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
27
|
Mohana Devi S, Mahalaxmi I, Kaavya J, Chinnkulandhai V, Balachandar V. Does epigenetics have a role in age related macular degeneration and diabetic retinopathy? Genes Dis 2020; 8:279-286. [PMID: 33997175 PMCID: PMC8093576 DOI: 10.1016/j.gendis.2020.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/06/2020] [Indexed: 02/08/2023] Open
Abstract
Epigenetic mechanisms play an important part in the regulation of gene expression and these alterations may induce long-term changes in gene function and metabolism. They have received extensive attention in bridging the gap between environmental exposures and disease development via their influence on gene expression. DNA methylation is the earliest discovered epigenetic alteration. In this review, we try to examine the role of DNA methylation and histone modification in Age related macular degeneration (AMD) and Diabetic Retinopathy (DR), its vascular complications and recent progress. Given the complex nature of AMD and DR, it is crucial to improve therapeutics which will greatly enhance the quality of life and reduce the burden for millions of patients living with these potentially blinding conditions.
Collapse
Affiliation(s)
- S Mohana Devi
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, 41/18, College Road, Chennai, 600006, India
| | - I Mahalaxmi
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Avinashilingam University for Women, Coimbatore, Tamil Nadu, 641046, India
| | - J Kaavya
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Avinashilingam University for Women, Coimbatore, Tamil Nadu, 641046, India
| | - V Chinnkulandhai
- Department of Biochemistry, Dr.N.G.P Arts and Science College, Coimbatore, Tamil Nadu, 641046, India
| | - V Balachandar
- Human Molecular Genetics and Stem Cells Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| |
Collapse
|
28
|
Yang DH, Lee SY. Diabetic kidney disease: seven questions. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2020. [DOI: 10.5124/jkma.2020.63.1.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Dong Ho Yang
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - So-Young Lee
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
29
|
Halladjian S, Miao H, Kouril D, Groller ME, Viola I, Isenberg T. Scale Trotter: Illustrative Visual Travels Across Negative Scales. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2020; 26:654-664. [PMID: 31425102 DOI: 10.1109/tvcg.2019.2934334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We present ScaleTrotter, a conceptual framework for an interactive, multi-scale visualization of biological mesoscale data and, specifically, genome data. ScaleTrotter allows viewers to smoothly transition from the nucleus of a cell to the atomistic composition of the DNA, while bridging several orders of magnitude in scale. The challenges in creating an interactive visualization of genome data are fundamentally different in several ways from those in other domains like astronomy that require a multi-scale representation as well. First, genome data has intertwined scale levels-the DNA is an extremely long, connected molecule that manifests itself at all scale levels. Second, elements of the DNA do not disappear as one zooms out-instead the scale levels at which they are observed group these elements differently. Third, we have detailed information and thus geometry for the entire dataset and for all scale levels, posing a challenge for interactive visual exploration. Finally, the conceptual scale levels for genome data are close in scale space, requiring us to find ways to visually embed a smaller scale into a coarser one. We address these challenges by creating a new multi-scale visualization concept. We use a scale-dependent camera model that controls the visual embedding of the scales into their respective parents, the rendering of a subset of the scale hierarchy, and the location, size, and scope of the view. In traversing the scales, ScaleTrotter is roaming between 2D and 3D visual representations that are depicted in integrated visuals. We discuss, specifically, how this form of multi-scale visualization follows from the specific characteristics of the genome data and describe its implementation. Finally, we discuss the implications of our work to the general illustrative depiction of multi-scale data.
Collapse
|
30
|
Low S, Zhang X, Wang J, Yeoh LY, Liu YL, Ang SF, Subramaniam T, Sum CF, Lim SC. Impact of haemoglobin A1c trajectories on chronic kidney disease progression in type 2 diabetes. Nephrology (Carlton) 2019; 24:1026-1032. [DOI: 10.1111/nep.13533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Serena Low
- Clinical Research UnitKhoo Teck Puat Hospital Yishun Singapore
| | - Xiao Zhang
- Clinical Research UnitKhoo Teck Puat Hospital Yishun Singapore
| | - Jiexun Wang
- Clinical Research UnitKhoo Teck Puat Hospital Yishun Singapore
| | - Lee Y Yeoh
- Department of MedicineSengkang General Hospital Singapore Singapore
| | - Yan L Liu
- Department of MedicineKhoo Teck Puat Hospital Yishun Singapore
| | - Su F Ang
- Clinical Research UnitKhoo Teck Puat Hospital Yishun Singapore
| | | | - Chee F Sum
- Diabetes CentreKhoo Teck Puat Hospital Singapore Singapore
| | - Su C Lim
- Clinical Research UnitKhoo Teck Puat Hospital Yishun Singapore
- Diabetes CentreKhoo Teck Puat Hospital Singapore Singapore
- Saw Swee Hock School of Public HealthNational University of Singapore Singapore Singapore
| |
Collapse
|
31
|
Wang H, Zhang Z, Sun J. The clinical evaluation of combined detection of microcirculation, lipid metabolism, and inflammatory-related factors in the treatment of diabetic nephropathy with atorvastatin. EUR J INFLAMM 2019. [DOI: 10.1177/2058739219847830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study was designed to analyze the effects of atorvastatin on microcirculation, blood lipids, inflammatory factors, and characteristic markers in patients with diabetic nephropathy. A total of 170 patients with diabetic nephropathy randomly divided into control and study groups with 85 patients in each group. The control group was treated with diet and lifestyle intervention, and hypoglycemic drugs. The study group was additionally treated with atorvastatin. Nitric oxide (NO), endothelin-1 (ET-1), thromboxane-2 (TXB2), 6-ketone-prostaglandin F-1α (6-Keto-PGF-1α), superoxide dismutase (SOD), total cholesterol (TC), triacylglycerols (TGs), low-density lipoprotein (LDL), high-density lipoprotein (HDL), C-reactive protein (CRP), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), homocysteine (Hcy), cystatin C (CysC), and vascular endothelial growth factor (VEGF) levels were observed for 8 weeks. Post-treatment of atorvastatin, the levels of NO, 6-Keto-PGF-1α, and SOD were significantly higher than pre-treatment in both groups, while the levels of ET-1 and TXB2 were lower than pre-treatment ( P < 0.05). The levels of NO, 6-Keto-PGF-1α, and SOD in the study group post-treatment were significantly higher ( P < 0.05) than the control group, and the levels of ET-1 and TXB2 in the study group were lower than the control group. After 8 weeks, the levels of TC, TG, and LDL were significantly lower, while the level of HDL was significantly higher in the study group. The level of TC was lower in the control group of post-treatment, while the HDL level was higher than pre-treatment ( P < 0.05). The levels of CRP, TNF-α, and IL-6 in the study group of post-treatment were significantly lower than pre-treatment comparing to the control group ( P < 0.05). There was no statistical significance ( P > 0.05) for above-mentioned indicators in control groups of pre- and post-treatment. The levels of VEGF, CysC, and Hcy in the two groups were lower than pre-treatment. Atorvastatin could effectively improve all the study parameters.
Collapse
Affiliation(s)
- Haicheng Wang
- Department of Nephrology, Linyi Central Hospital, Linyi, P.R. China
| | - Zhengfang Zhang
- Department of Nephrology, Linyi Central Hospital, Linyi, P.R. China
| | - Jiali Sun
- Department of Nephrology, Haiyang People’s Hospital, Haiyang, P.R. China
| |
Collapse
|
32
|
Samblas M, Milagro FI, Martínez A. DNA methylation markers in obesity, metabolic syndrome, and weight loss. Epigenetics 2019; 14:421-444. [PMID: 30915894 DOI: 10.1080/15592294.2019.1595297] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The fact that not all individuals exposed to the same environmental risk factors develop obesity supports the hypothesis of the existence of underlying genetic and epigenetic elements. There is suggestive evidence that environmental stimuli, such as dietary pattern, particularly during pregnancy and early life, but also in adult life, can induce changes in DNA methylation predisposing to obesity and related comorbidities. In this context, the DNA methylation marks of each individual have emerged not only as a promising tool for the prediction, screening, diagnosis, and prognosis of obesity and metabolic syndrome features, but also for the improvement of weight loss therapies in the context of precision nutrition. The main objectives in this field are to understand the mechanisms involved in transgenerational epigenetic inheritance, and featuring the nutritional and lifestyle factors implicated in the epigenetic modifications. Likewise, DNA methylation modulation caused by diet and environment may be a target for newer therapeutic strategies concerning the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Mirian Samblas
- a Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research , University of Navarra , Pamplona , Spain
| | - Fermín I Milagro
- a Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research , University of Navarra , Pamplona , Spain.,b CIBERobn, CIBER Fisiopatología de la Obesidad y Nutrición , Instituto de Salud Carlos III. Madrid , Spain.,c IdiSNA, Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona , Spain
| | - Alfredo Martínez
- a Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research , University of Navarra , Pamplona , Spain.,b CIBERobn, CIBER Fisiopatología de la Obesidad y Nutrición , Instituto de Salud Carlos III. Madrid , Spain.,c IdiSNA, Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona , Spain.,d IMDEA, Research Institute on Food & Health Sciences , Madrid , Spain
| |
Collapse
|
33
|
Glycine Suppresses AGE/RAGE Signaling Pathway and Subsequent Oxidative Stress by Restoring Glo1 Function in the Aorta of Diabetic Rats and in HUVECs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4628962. [PMID: 30944692 PMCID: PMC6421782 DOI: 10.1155/2019/4628962] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/13/2019] [Indexed: 12/20/2022]
Abstract
Oxidative stress plays a crucial role in the pathogenesis of diabetic vascular complications. It is known that the accumulation of advanced glycation end products (AGEs) and the activation of the receptor of AGEs (RAGE) induce sustained oxidative stress in the vascular tissue. Growing evidence indicates that glycine, the simplest amino acid, exerts antioxidant and antiglycation effects and also improves vascular function. However, the mechanism whereby glycine protects vascular tissue against oxidative stress in models with diabetes has not been investigated. In the present study, we evaluated whether glycine can attenuate oxidative stress by suppressing the AGE/RAGE signaling pathway in the aorta of streptozotocin-induced diabetic rats and in human umbilical vascular endothelial cells (HUVECs). Our results showed that oral glycine administration increased NO content and ameliorated oxidative stress in the serum and aorta of diabetic rats. The AGE/RAGE signaling pathway in the aorta of diabetic rats was significantly attenuated by glycine treatment as manifested by decreases in levels of AGEs, RAGE, Nox4, and NF-κB p65. The suppressive effect of glycine on the formation of AGEs was associated with increased activity and expression of aortic glyoxalase-1 (Glo1), a crucial enzyme that degrades methylglyoxal (MG), the major precursor of AGEs. In MG-treated HUVECs, glycine restored the function of Glo1, suppressed the AGE/RAGE signaling pathway, and inhibited the generation of reactive oxygen species. In addition, the reduction in the formation of AGEs in HUVECs caused by glycine treatment was inhibited by Glo1 inhibition. Taken together, our study provides evidence that glycine might inhibit the AGE/RAGE pathway and subsequent oxidative stress by improving Glo1 function, thus protecting against diabetic macrovascular complications.
Collapse
|
34
|
Modafferi S, Ries M, Calabrese V, Schmitt CP, Nawroth P, Kopf S, Peters V. Clinical Trials on Diabetic Nephropathy: A Cross-Sectional Analysis. Diabetes Ther 2019; 10:229-243. [PMID: 30617943 PMCID: PMC6349284 DOI: 10.1007/s13300-018-0551-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Treatment options and decisions are often based on the results of clinical trials. We have evaluated the public availability of results from completed, registered phase III clinical trials on diabetic nephropathy and current treatment options. METHODS This was a cross-sectional analysis in which STrengthening the Reporting of OBservational studies in Epidemiology criteria were applied for design and analysis. In June 2017, 34 completed phase III clinical trials on diabetic nephropathy in the ClinicalTrials. gov registry were identified and matched to publications in the ClinicalTrials.gov registry and to those in the PubMed and Google Scholar databases. If no publication was identified, the principal investigator was contacted. The ratio of published and non-published studies was calculated. Various parameters, including study design, drugs, and comparators provided, were analyzed. RESULTS Drugs/supplements belonged to 26 different categories of medications, with the main ones being angiotensin-converting enzyme inhibitors, angiotensin-II receptors blockers, and dipeptidyl-peptidase-4-inhibitors. Among the trials completed before 2016 (n = 32), 22 (69%) were published, and ten (31%) remained unpublished. Thus, data on 11 different interventions and more than 1000 patients remained undisclosed. Mean time to publication was 26.5 months, which is longer than the time constrictions imposed by the U.S. Food and Drug Administration Amendments Act. Most trials only showed weak effects on micro- and macroalbuminuria, with an absolute risk reduction of 1.0 and 0.3%, respectively, and the number needed to treat varied between 91 and 333, without any relevant effect on end-stage-renal disease by intensive glucose-lowering treatment. Comparison of the results, however, was difficult since study design, interventions, and the renal outcome parameters vary greatly between the studies. CONCLUSION Despite the financial and human resources involved and the relevance for therapeutic guidelines and clinical decisions, about one-third of phase III clinical trials on diabetic nephropathy remain unpublished. Interventions used in published trials showed a low efficacy on renal outcome. FUNDING Deutsche Forschungsgemeinschaft (DFG): SFB 1118.
Collapse
Affiliation(s)
- Sergio Modafferi
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Markus Ries
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Claus P Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Peter Nawroth
- Department of Endocrinology, Diabetology and Clinical Chemistry, University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
- Deutsches Zentrum für Diabetesforschung e.V. (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Institute for Diabetes and Cancer, Helmholtz Zentrum, Neuherberg, Germany
| | - Stefan Kopf
- Department of Endocrinology, Diabetology and Clinical Chemistry, University Hospital Heidelberg, University Heidelberg, Heidelberg, Germany
- Deutsches Zentrum für Diabetesforschung e.V. (DZD), Neuherberg, Germany
| | - Verena Peters
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
35
|
Willemsen L, Neele AE, van der Velden S, Prange KHM, den Toom M, van Roomen CPAA, Reiche ME, Griffith GR, Gijbels MJJ, Lutgens E, de Winther MPJ. Peritoneal macrophages have an impaired immune response in obesity which can be reversed by subsequent weight loss. BMJ Open Diabetes Res Care 2019; 7:e000751. [PMID: 31798899 PMCID: PMC6861071 DOI: 10.1136/bmjdrc-2019-000751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/24/2019] [Accepted: 10/14/2019] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Obesity is recognized as a risk factor for various microbial infections. The immune system, which is affected by obesity, plays an important role in the pathophysiology of these infections and other obesity-related comorbidities. Weight loss is considered the most obvious treatment for obesity. However, multiple studies suggest that the comorbidities of obesity may persist after weight loss. Deregulation of immune cells including adipose tissue macrophages of obese individuals has been extensively studied, but how obesity and subsequent weight loss affect immune cell function outside adipose tissue is not well defined. RESEARCH DESIGN AND METHODS Here we investigated the phenotype of non-adipose tissue macrophages by transcriptional characterization of thioglycollate-elicited peritoneal macrophages (PM) from mice with diet-induced obesity and type 2 diabetes (T2D). Subsequently, we defined the characteristics of PMs after weight loss and mimicked a bacterial infection by exposing PMs to lipopolysaccharide. RESULTS AND CONCLUSIONS In contrast to the proinflammatory phenotype of adipose tissue macrophages in obesity and T2D, we found a deactivated state of PMs in obesity and T2D. Weight loss could reverse this deactivated macrophage phenotype. Anti-inflammatory characteristics of these non-adipose macrophages may explain why patients with obesity and T2D have an impaired immune response against pathogens. Our data also suggest that losing weight restores macrophage function and thus contributes to the reduction of immune-related comorbidities in patients.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/therapy
- Diet, High-Fat
- Dietary Fats/pharmacology
- Immunity, Cellular/drug effects
- Immunity, Cellular/physiology
- Insulin Resistance/physiology
- Macrophage Activation/drug effects
- Macrophage Activation/physiology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Obesity/complications
- Obesity/immunology
- Obesity/pathology
- Obesity/therapy
- Weight Loss/immunology
- Weight Loss/physiology
Collapse
Affiliation(s)
- Lisa Willemsen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Annette E Neele
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Saskia van der Velden
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Koen H M Prange
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Myrthe den Toom
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Cindy P A A van Roomen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Myrthe E Reiche
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Guillermo R Griffith
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marion J J Gijbels
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Departments of Pathology and Molecular Genetics, CARIM School for Cardiovascular Diseases and GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University Munich, Munich, Germany
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC-Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University Munich, Munich, Germany
| |
Collapse
|
36
|
Hurtado Del Pozo C, Garreta E, Izpisúa Belmonte JC, Montserrat N. Modeling epigenetic modifications in renal development and disease with organoids and genome editing. Dis Model Mech 2018; 11:dmm035048. [PMID: 30459215 PMCID: PMC6262817 DOI: 10.1242/dmm.035048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Understanding epigenetic mechanisms is crucial to our comprehension of gene regulation in development and disease. In the past decades, different studies have shown the role of epigenetic modifications and modifiers in renal disease, especially during its progression towards chronic and end-stage renal disease. Thus, the identification of genetic variation associated with chronic kidney disease has resulted in better clinical management of patients. Despite the importance of these findings, the translation of genotype-phenotype data into gene-based medicine in chronic kidney disease populations still lacks faithful cellular or animal models that recapitulate the key aspects of the human kidney. The latest advances in the field of stem cells have shown that it is possible to emulate kidney development and function with organoids derived from human pluripotent stem cells. These have successfully recapitulated not only kidney differentiation, but also the specific phenotypical traits related to kidney function. The combination of this methodology with CRISPR/Cas9 genome editing has already helped researchers to model different genetic kidney disorders. Nowadays, CRISPR/Cas9-based approaches also allow epigenetic modifications, and thus represent an unprecedented tool for the screening of genetic variants, epigenetic modifications or even changes in chromatin structure that are altered in renal disease. In this Review, we discuss these technical advances in kidney modeling, and offer an overview of the role of epigenetic regulation in kidney development and disease.
Collapse
Affiliation(s)
- Carmen Hurtado Del Pozo
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| | - Elena Garreta
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| | | | - Nuria Montserrat
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| |
Collapse
|
37
|
Cerulus B, Jariani A, Perez-Samper G, Vermeersch L, Pietsch JMJ, Crane MM, New AM, Gallone B, Roncoroni M, Dzialo MC, Govers SK, Hendrickx JO, Galle E, Coomans M, Berden P, Verbandt S, Swain PS, Verstrepen KJ. Transition between fermentation and respiration determines history-dependent behavior in fluctuating carbon sources. eLife 2018; 7:e39234. [PMID: 30299256 PMCID: PMC6211830 DOI: 10.7554/elife.39234] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/05/2018] [Indexed: 01/24/2023] Open
Abstract
Cells constantly adapt to environmental fluctuations. These physiological changes require time and therefore cause a lag phase during which the cells do not function optimally. Interestingly, past exposure to an environmental condition can shorten the time needed to adapt when the condition re-occurs, even in daughter cells that never directly encountered the initial condition. Here, we use the molecular toolbox of Saccharomyces cerevisiae to systematically unravel the molecular mechanism underlying such history-dependent behavior in transitions between glucose and maltose. In contrast to previous hypotheses, the behavior does not depend on persistence of proteins involved in metabolism of a specific sugar. Instead, presence of glucose induces a gradual decline in the cells' ability to activate respiration, which is needed to metabolize alternative carbon sources. These results reveal how trans-generational transitions in central carbon metabolism generate history-dependent behavior in yeast, and provide a mechanistic framework for similar phenomena in other cell types.
Collapse
Affiliation(s)
- Bram Cerulus
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Abbas Jariani
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Gemma Perez-Samper
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Lieselotte Vermeersch
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Julian MJ Pietsch
- Centre for Synthetic and Systems Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Matthew M Crane
- Centre for Synthetic and Systems Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
- Department of PathologyUniversity of WashingtonWashingtonUnited States
| | - Aaron M New
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Brigida Gallone
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
- Department of Plant Biotechnology and BioinformaticsGhent UniversityGhentBelgium
- VIB Center for Plant Systems BiologyGhentBelgium
| | - Miguel Roncoroni
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Maria C Dzialo
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Sander K Govers
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Jhana O Hendrickx
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Eva Galle
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Maarten Coomans
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Pieter Berden
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Sara Verbandt
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| | - Peter S Swain
- Centre for Synthetic and Systems Biology, School of Biological SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Kevin J Verstrepen
- VIB Laboratory for Systems BiologyVIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Departement Microbiële en Moleculaire Systemen (M2S)CMPG Laboratory of Genetics and GenomicsLeuvenBelgium
| |
Collapse
|
38
|
Viola I, Isenberg T. Pondering the Concept of Abstraction in (Illustrative) Visualization. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2018; 24:2573-2588. [PMID: 28880182 DOI: 10.1109/tvcg.2017.2747545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We explore the concept of abstraction as it is used in visualization, with the ultimate goal of understanding and formally defining it. Researchers so far have used the concept of abstraction largely by intuition without a precise meaning. This lack of specificity left questions on the characteristics of abstraction, its variants, its control, or its ultimate potential for visualization and, in particular, illustrative visualization mostly unanswered. In this paper we thus provide a first formalization of the abstraction concept and discuss how this formalization affects the application of abstraction in a variety of visualization scenarios. Based on this discussion, we derive a number of open questions still waiting to be answered, thus formulating a research agenda for the use of abstraction for the visual representation and exploration of data. This paper, therefore, is intended to provide a contribution to the discussion of the theoretical foundations of our field, rather than attempting to provide a completed and final theory.
Collapse
|
39
|
Alicic RZ, Rooney MT, Tuttle KR. Diabetic Kidney Disease: Challenges, Progress, and Possibilities. Clin J Am Soc Nephrol 2017; 12:2032-2045. [PMID: 28522654 PMCID: PMC5718284 DOI: 10.2215/cjn.11491116] [Citation(s) in RCA: 1731] [Impact Index Per Article: 216.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic kidney disease develops in approximately 40% of patients who are diabetic and is the leading cause of CKD worldwide. Although ESRD may be the most recognizable consequence of diabetic kidney disease, the majority of patients actually die from cardiovascular diseases and infections before needing kidney replacement therapy. The natural history of diabetic kidney disease includes glomerular hyperfiltration, progressive albuminuria, declining GFR, and ultimately, ESRD. Metabolic changes associated with diabetes lead to glomerular hypertrophy, glomerulosclerosis, and tubulointerstitial inflammation and fibrosis. Despite current therapies, there is large residual risk of diabetic kidney disease onset and progression. Therefore, widespread innovation is urgently needed to improve health outcomes for patients with diabetic kidney disease. Achieving this goal will require characterization of new biomarkers, designing clinical trials that evaluate clinically pertinent end points, and development of therapeutic agents targeting kidney-specific disease mechanisms (e.g., glomerular hyperfiltration, inflammation, and fibrosis). Additionally, greater attention to dissemination and implementation of best practices is needed in both clinical and community settings. INTRODUCTION
Collapse
Affiliation(s)
- Radica Z. Alicic
- Providence Health Care, Spokane, Washington
- University of Washington School of Medicine, Seattle, Washington
| | | | - Katherine R. Tuttle
- Providence Health Care, Spokane, Washington
- University of Washington School of Medicine, Seattle, Washington
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington
- Institute of Translational Health Sciences, Seattle, Washington; and
- Kidney Research Institute, Seattle, Washington
| |
Collapse
|
40
|
Low S, Lim SC, Yeoh LY, Liu YL, Liu JJ, Fun S, Su C, Zhang X, Subramaniam T, Sum CF. Effect of long-term glycemic variability on estimated glomerular filtration rate decline among patients with type 2 diabetes mellitus: Insights from the Diabetic Nephropathy Cohort in Singapore. J Diabetes 2017; 9:908-919. [PMID: 27935225 DOI: 10.1111/1753-0407.12512] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/06/2016] [Accepted: 11/25/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In the present study, we examined the association between HbA1c variability and renal disease progression based on estimated glomerular filtration rate (eGFR) decline in patients with type 2 diabetes mellitus (T2DM) in Singapore. METHODS Glycemic burden and renal function were retrospectively assessed in 1628 patients in 2002-2014. Multivariable logistic regression was used to assess the relationships between HbA1c variability (expressed as HbA1c coefficient of variation [HbA1c-CV] in quartiles), HbA1c intrapersonal mean (HbA1c-IM), and eGFR decline, adjusted for baseline covariates. RESULTS Among patients with relatively good glycemic control (i.e. HbA1c-IM below the median cohort value [8.0%]), HbA1c-CV Quartile 4 was associated with eGFR decline (odds ratio [OR] 1.88; 95% confidence interval [CI] 1.10-3.25). The OR for HbA1c-CV Quartile 4 was 2.20 (95% CI 1.24-3.89) after additional adjustment for HbA1c-IM. Where HbA1c-IM was above the median cohort value, HbA1c-CV Quartiles 3 and 4 were associated with eGFR decline, with ORs of 2.60 (95% CI 1.48-4.55) and 3.29 (95% CI 1.89-5.76) respectively. After further adjusting for HbA1c-IM, the ORs for Quartiles 3 and 4 were 2.69 (95% CI 1.53-4.74) and 3.51 (95% CI 1.98-6.21), respectively. CONCLUSIONS Variability in HbA1c is strongly and independently associated with eGFR decline in patients with T2DM independent of mean HbA1c. The findings may highlight the importance of sustained stable glycemic control in management of diabetes mellitus.
Collapse
Affiliation(s)
- Serena Low
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Su C Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
- Diabetes Centre, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Lee Y Yeoh
- Department of Medicine, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Yan L Liu
- Department of Medicine, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Jian J Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Sharon Fun
- Diabetes Centre, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Chang Su
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Xiao Zhang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | | | - Chee F Sum
- Diabetes Centre, Khoo Teck Puat Hospital, Singapore, Singapore
| |
Collapse
|
41
|
Zhang L, Zhang Q, Liu S, Chen Y, Li R, Lin T, Yu C, Zhang H, Huang Z, Zhao X, Tan X, Li Z, Ye Z, Ma J, Zhang B, Wang W, Shi W, Liang X. DNA methyltransferase 1 may be a therapy target for attenuating diabetic nephropathy and podocyte injury. Kidney Int 2017; 92:140-153. [PMID: 28318634 DOI: 10.1016/j.kint.2017.01.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/21/2016] [Accepted: 01/05/2017] [Indexed: 01/19/2023]
Abstract
The contribution of DNA methylation to diabetic nephropathy, especially the effect on podocyte integrity, is not clarified. Here we found that albuminuria in a db/db mouse model was markedly attenuated after treatment with a DNA methylation inhibitor. This was accompanied by alleviation of glomerular hypertrophy, mesangial matrix expansion, and podocyte injury. The expression of DNA methyltransferase 1 (Dnmt1), nuclear factor Sp1, and nuclear factor kappa B (NFκB)-p65 markedly increased in podocytes in vivo and in vitro under the diabetic state. The increased expression of Dnmt1 was attenuated after treatment with 5-azacytidine or 5-aza-2'-deoxycytidine or Dnmt1 knockdown, accompanied by restored decreased podocyte slit diaphragm proteins resulting from hypermethylation and improved podocyte motility. Further studies found that increased Sp1 and NFκB-p65 interacted in the nucleus of podocytes incubated with high glucose, and Sp1 bound to the Dnmt1 promoter region. The involvement of the Sp1/NFκB-p65 complex in Dnmt1 regulation was confirmed by the observation that Sp1 knockdown using mithramycin A or siRNA decreased Dnmt1 protein levels. The luciferase reporter assay further indicated that Dnmt1 was a direct target of Sp1. Thus, inhibition of DNA methylation may be a new therapeutic avenue for treating diabetic nephropathy. Hence, the Sp1/NFκB p65-Dnmt1 pathway may be exploited as a therapeutic target for protecting against podocyte injury in diabetic nephropathy.
Collapse
Affiliation(s)
- Li Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Shuangxin Liu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuanhan Chen
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ruizhao Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ting Lin
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chunping Yu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hong Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhongshun Huang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinchen Zhao
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Southern Medical University, Guangzhou, China
| | - Xiaofan Tan
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhuo Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhiming Ye
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jianchao Ma
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bin Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wenjian Wang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wei Shi
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Xinling Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
42
|
Sharma D, Bhattacharya P, Kalia K, Tiwari V. Diabetic nephropathy: New insights into established therapeutic paradigms and novel molecular targets. Diabetes Res Clin Pract 2017; 128:91-108. [PMID: 28453961 DOI: 10.1016/j.diabres.2017.04.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/07/2017] [Indexed: 02/06/2023]
Abstract
Diabetic nephropathy is one of the most prevalent microvascular complication in patients suffering from diabetes and is reported to be the major cause of renal failure when compared to any other kidney disease. Currently, available therapies provide only symptomatic relief and unable to treat the underlying pathophysiology of diabetic nephropathy. This review will explore new insights into the established therapeutic paradigms targeting oxidative stress, inflammation and endoplasmic reticulum stress with the focus on recent clinical developments. Apart from this, the involvement of novel cellular and molecular mechanisms including the role of endothelin-receptor antagonists, Wnt signaling pathway, epigenetics and micro RNA is also discussed so that key molecular switches involved in the pathogenesis of diabetic nephropathy can be identified. Elucidating new molecular pathways will help in the development of novel therapeutics for the prevention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Dilip Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India.
| | - Vinod Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India.
| |
Collapse
|
43
|
Lu Z, Liu N, Wang F. Epigenetic Regulations in Diabetic Nephropathy. J Diabetes Res 2017; 2017:7805058. [PMID: 28401169 PMCID: PMC5376412 DOI: 10.1155/2017/7805058] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/10/2023] Open
Abstract
Diabetic nephropathy (DN) is a chronic complication of diabetes and the most common cause of end-stage kidney disease. It has been reported that multiple factors are involved in the pathogenesis of DN, while the molecular mechanisms that lead to DN are still not fully understood. Numerous risk factors for the development of diabetic nephropathy have been proposed, including ethnicity and inherited genetic differences. Recently, with the development of high-throughput technologies, there is emerging evidence that suggests the important role of epigenetic mechanisms in the pathogenesis of DN. Epigenetic regulations, including DNA methylation, noncoding RNAs, and histone modifications, play a pivotal role in DN pathogenesis by a second layer of gene regulation. All these findings can contribute to developing novel therapies for DN.
Collapse
Affiliation(s)
- Zeyuan Lu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
44
|
Novel insights into DNA methylation and its critical implications in diabetic vascular complications. Biosci Rep 2017; 37:BSR20160611. [PMID: 28183874 PMCID: PMC5350598 DOI: 10.1042/bsr20160611] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 12/23/2022] Open
Abstract
Recent epidemiological and clinical studies have shown that type 2 diabetic patients can develop diabetic vascular complications even after intensive glycaemic control. It has been suggested that this phenomenon could be explained by the hypothesis of 'metabolic memory'. The underlying mechanisms between these enduring effects and the prior hyperglycaemic state are still not well understood. Preliminary studies demonstrate that hyperglycaemia can regulate gene expression by epigenetic modifications, such as DNA methylation, which can persistently exist even after glucose normalization. Increasing evidence shows that epigenetic mechanisms may play a substantial role in the pathophysiology of diabetes and its associated vascular complications, including atherosclerosis, diabetic cardiomyopathy (DCM), nephropathy and retinopathy. In this review, we will examine the growing role of DNA methylation in diabetes and its vascular complications, thus it can provide critical implications for the early prevention of diabetes and its vascular complications.
Collapse
|
45
|
Li W, Chen X, Lin M, Huang D. Up-regulated HOTAIR induced by fatty acids inhibits PTEN expression and increases triglycerides accumulation in HepG2 cells. Food Nutr Res 2017; 61:1412794. [DOI: 10.1080/16546628.2017.1412794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Weiping Li
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medcial College, Shantou, China
| | - Xiaoge Chen
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medcial College, Shantou, China
| | - Miaozhi Lin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medcial College, Shantou, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics and Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, China
| |
Collapse
|
46
|
Sun J, Wang Y, Cui W, Lou Y, Sun G, Zhang D, Miao L. Role of Epigenetic Histone Modifications in Diabetic Kidney Disease Involving Renal Fibrosis. J Diabetes Res 2017; 2017:7242384. [PMID: 28695133 PMCID: PMC5485509 DOI: 10.1155/2017/7242384] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/14/2017] [Indexed: 12/18/2022] Open
Abstract
One of the commonest causes of end-stage renal disease is diabetic kidney disease (DKD). Renal fibrosis, characterized by the accumulation of extracellular matrix (ECM) proteins in glomerular basement membranes and the tubulointerstitium, is the final manifestation of DKD. The TGF-β pathway triggers epithelial-to-mesenchymal transition (EMT), which plays a key role in the accumulation of ECM proteins in DKD. DCCT/EDIC studies have shown that DKD often persists and progresses despite glycemic control in diabetes once DKD sets in due to prior exposure to hyperglycemia called "metabolic memory." These imply that epigenetic factors modulate kidney gene expression. There is evidence to suggest that in diabetes and hyperglycemia, epigenetic histone modifications have a significant effect in modulating renal fibrotic and ECM gene expression induced by TGF-β1, as well as its downstream profibrotic genes. Histone modifications are also implicated in renal fibrosis through its ability to regulate the EMT process triggered by TGF-β signaling. In view of this, efforts are being made to develop HAT, HDAC, and HMT inhibitors to delay, stop, or even reverse DKD. In this review, we outline the latest advances that are being made to regulate histone modifications involved in DKD.
Collapse
Affiliation(s)
- Jing Sun
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Yangwei Wang
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Yan Lou
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Guangdong Sun
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Dongmei Zhang
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Lining Miao
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
- *Lining Miao:
| |
Collapse
|
47
|
Tikoo K, Sharma E, Amara VR, Pamulapati H, Dhawale VS. Metformin Improves Metabolic Memory in High Fat Diet (HFD)-induced Renal Dysfunction. J Biol Chem 2016; 291:21848-21856. [PMID: 27551045 DOI: 10.1074/jbc.c116.732990] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/04/2016] [Indexed: 12/14/2022] Open
Abstract
Recently, we have shown that high fat diet (HFD) in vivo and in vitro generates metabolic memory by altering H3K36me2 and H3K27me3 on the promoter of FOXO1 (transcription factor of gluconeogenic genes) (Kumar, S., Pamulapati, H., and Tikoo, K. (2016) Mol. Cell. Endocrinol. 422, 233-242). Here we checked the hypothesis whether concomitant diet reversal and metformin could overcome HFD-induced metabolic memory and renal damage. Male adult Sprague-Dawley rats were rendered insulin-resistant by feeding high fat diet for 16 weeks. Then the rats were subjected to diet reversal alone and along with metformin for 8 weeks. Biochemical and histological markers of insulin resistance and kidney function were measured. Blood pressure and in vivo vascular reactivity to angiotensin II (200 ng kg-1) were also checked. Diet reversal could improve lipid profile but could not prevent renal complications induced by HFD. Interestingly, metformin along with diet reversal restored the levels of blood glucose, triglycerides, cholesterol, blood urea nitrogen, and creatinine. In kidney, metformin increased the activation of AMP-activated protein kinase (AMPK) and decreased inflammatory markers (COX-2 and IL-1β) and apoptotic markers (poly(ADP-ribose) polymerase (PARP) and caspase 3). Metformin was effective in lowering elevated basal blood pressure and acute change in mean arterial pressure in response to angiotensin II (Ang II). It also attenuated tubulointerstitial fibrosis and glomerulosclerosis induced by HFD feeding in kidney. Here we report, for the first time, that metformin treatment overcomes metabolic memory and prevents HFD-induced renal damage.
Collapse
Affiliation(s)
- Kulbhushan Tikoo
- From the Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Ekta Sharma
- From the Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Venkateswara Rao Amara
- From the Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Himani Pamulapati
- From the Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Vaibhav Shrirang Dhawale
- From the Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| |
Collapse
|
48
|
Zhang H, Li A, Zhang W, Huang Z, Wang J, Yi B. High glucose-induced cytoplasmic translocation of Dnmt3a contributes to CTGF hypo-methylation in mesangial cells. Biosci Rep 2016; 36:e00362. [PMID: 27364355 PMCID: PMC4974599 DOI: 10.1042/bsr20160141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 06/13/2016] [Accepted: 06/28/2016] [Indexed: 01/16/2023] Open
Abstract
Connective tissue growth factor (CTGF) plays an essential role in the pathogenesis of diabetic nephropathy and we have previously identified that high glucose induced the expression of CTGF by decreasing DNA methylation. The aim of the present study was to investigate the underlying mechanisms of the high glucose-induced CTGF hypo-methylation. Human glomerular mesangial cells (hMSCs) were treated with low glucose (5 mM), mannitol (30 mM) or high glucose (30 mM) respectively. Immunofluorescence staining, real-time quantitative PCR and western blotting were performed to determine the subcellular distribution and expression of CTGF and Dnmt3a. ChIP-PCR assay was applied to investigate the capability of Dnmt3a to bind the CpG island of CTGF. Our results showed that high glucose induced both mRNA and protein expressions of CTGF, and led to increased cytoplasmic translocation of Dnmt3a in cultured hMSCs. The nuclear Dnmt3a protein was significantly reduced after high glucose treatment, although the expression of total Dnmt3a protein was not altered. We further discovered that ERK/MAPK signalling contributed to the high glucose-induced cytoplasmic translocation of Dnmt3a. Consequently, less Dnmt3a protein was bound to the CpG island of CTGF promoter, which induced an increase in CTGF expression by epigenetic regulation in the presence of high glucose. In conclusion, high glucose induces cytoplasmic translocation of Dnmt3a, possibly through activating ERK/MAPK signalling pathway, which contributes to the decreased binding of Dnmt3a on CTGF promoter and the subsequent CTGF hypo-methylation in diabetic nephropathy.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Aimei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhijun Huang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Jianwen Wang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Bin Yi
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
49
|
Inagi R. RAGE and glyoxalase in kidney disease. Glycoconj J 2016; 33:619-26. [PMID: 27270765 DOI: 10.1007/s10719-016-9689-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 12/19/2022]
Abstract
Glycation is an important reaction in the regulation of physiological state. When poorly controlled, however, glycation can also result in the accumulation of glycated proteins (advanced glycation endproducts; AGEs) in the body. This AGE accumulation is termed glycative stress, and is an established pathological factor: to date, glycative stress has been closely associated with not only kidney diseases, but also kidney aging. Accumulating evidence demonstrates that the progression of renal tubular damage and tubular aging are often correlated with activation of the receptor for the AGE (RAGE)-AGE pathway or decreased activity of glyoxalase 1, which is an anti-glycation enzyme to lower glycative stress. Further, glycative stress exacerbates the derangement of protein homeostasis: the posttranslationally modified proteins by glycation often lose or gain their functions. Such deranged protein homeostasis leads to endoplasmic reticulum (ER) stress, a state of ER dysfunction in which the quality control of proteins is defective, as well as to induction of its stress signal, the unfolded protein response (UPR), in the kidney. The lowering of glycative stress via modulation of RAGE-AGE axis or glyoxalase 1 activity is beneficial for tubular homeostasis and the subsequent prevention and treatment of kidney disease, suggesting the possibility of novel therapeutic approaches which target glycative stress. In this review, we focused on the impact of glycative stress in the kidney, especially the role of RAGE and glyoxalase 1. Further we also discuss the crosstalk between glycative stress and ER stress in their effect on protein homeostasis.
Collapse
Affiliation(s)
- Reiko Inagi
- Division of Chronic Kidney Disease (CKD) Pathophysiology, The University of Tokyo Graduate School of Medicine, 7-3-1, Hongo Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
50
|
Yamada-Obara N, Yamagishi SI, Taguchi K, Kaida Y, Yokoro M, Nakayama Y, Ando R, Asanuma K, Matsui T, Ueda S, Okuda S, Fukami K. Maternal exposure to high-fat and high-fructose diet evokes hypoadiponectinemia and kidney injury in rat offspring. Clin Exp Nephrol 2016; 20:853-861. [PMID: 27179663 DOI: 10.1007/s10157-016-1265-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/27/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Maternal exposure to overnutrition during fetal development contributes to metabolic and renal damage in offspring. Adiponectin plays a protective role against obesity-related renal injury. However, role of adiponectin in renal injury of offspring exposed to maternal overnutrition remains unknown. We addressed the issue. METHODS Female Sprague-Dawley rats were fed either a standard (N) or a high-fat and high-fructose (HFF)-diet for 6 weeks before mating, and kept each diet during the gestation and lactation period. After 4 weeks postpartum, all the offspring were fed N diet, and followed by 12 weeks. Kidney weight, urinary albumin excretion, blood pressure, and blood chemistry, including adiponectin and malondialdehyde, a marker of oxidative stress, were evaluated in the offspring. RESULTS Compared with N-offspring, serum adiponectin levels of 1-day- and 4-week-old HFF-offspring were significantly lower, the latter of which was inversely associated with malondialdehyde. Kidney weight was significantly decreased in 1-day-old HFF-offspring, whereas increased in 4-week-old HFF-offspring. Urinary albumin excretion levels of HFF-offspring at 8, 12, and 16-week old were significantly higher than those of N-offspring at the same age, whose levels at 16-week old were inversely correlated with plasma adiponectin. Compared with N-offspring, HFF-offspring at 16-week old exhibited glomerulosclerosis, hyperglycemia, and high mean blood pressure associated with reduced podocin and increased transforming growth factor-β1 expression in the kidneys. CONCLUSIONS Our present study suggests that exposure to maternal HFF-diet during fetal and early postnatal development induces hypoadiponectinemia in offspring, which might cause renal injury and metabolic derangements later in life.
Collapse
Affiliation(s)
- Nana Yamada-Obara
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Kensei Taguchi
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Yusuke Kaida
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Miyuki Yokoro
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Yosuke Nakayama
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Ryotaro Ando
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Katsuhiko Asanuma
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Seiji Ueda
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Seiya Okuda
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan
| | - Kei Fukami
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Fukuoka, Japan.
| |
Collapse
|