1
|
de Lorenzo A, Bomback AS, Mihic N. High Protein Diets and Glomerular Hyperfiltration in Athletes and Bodybuilders: Is Chronic Kidney Disease the Real Finish Line? Sports Med 2024; 54:2481-2495. [PMID: 39196487 DOI: 10.1007/s40279-024-02086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/29/2024]
Abstract
Several observational and experimental studies in humans have suggested that high protein intake (PI) causes intraglomerular hypertension leading to hyperfiltration. This phenomenon results in progressive loss of renal function with long-term exposure to high-protein diets (HPDs), even in healthy people. The recommended daily allowance for PI is 0.83 g/kg per day, which meets the protein requirement for approximately 98% of the population. A HPD is defined as a protein consumption > 1.5 g/kg per day. Athletes and bodybuilders are encouraged to follow HPDs to optimize muscle protein balance, increase lean body mass, and enhance performance. A series of studies in resistance-trained athletes looking at HPD has been published concluding that there are no harmful effects of HPD on renal health. However, the aim of these studies was to evaluate body composition changes and they were not designed to assess safety or kidney outcomes. Here we review the effects of HPD on kidney health in athletes and healthy individuals with normal kidney function.
Collapse
Affiliation(s)
- Alberto de Lorenzo
- Department of Nephrology, Hospital Universitario HM Sanchinarro, Madrid, Spain.
- Department of Nephrology, Hospital Universitario de Getafe, Universidad Europea de Madrid, Madrid, Spain.
| | - Andrew S Bomback
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, Presbyterian Hospital, New York, USA
| | - Niko Mihic
- Chief Medical Officer of Real Madrid CF, Madrid, Spain
| |
Collapse
|
2
|
Ding W, Yang X, Lai K, Jiang Y, Liu Y. The potential of therapeutic strategies targeting mitochondrial biogenesis for the treatment of insulin resistance and type 2 diabetes mellitus. Arch Pharm Res 2024; 47:219-248. [PMID: 38485900 DOI: 10.1007/s12272-024-01490-5] [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: 12/10/2023] [Accepted: 03/07/2024] [Indexed: 04/07/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a persistent metabolic disorder marked by deficiencies in insulin secretion and/or function, affecting various tissues and organs and leading to numerous complications. Mitochondrial biogenesis, the process by which cells generate new mitochondria utilizing existing ones plays a crucial role in energy homeostasis, glucose metabolism, and lipid handling. Recent evidence suggests that promoting mitochondrial biogenesis can alleviate insulin resistance in the liver, adipose tissue, and skeletal muscle while improving pancreatic β-cell function. Moreover, enhanced mitochondrial biogenesis has been shown to ameliorate T2DM symptoms and may contribute to therapeutic effects for the treatment of diabetic nephropathy, cardiomyopathy, retinopathy, and neuropathy. This review summarizes the intricate connection between mitochondrial biogenesis and T2DM, highlighting the potential of novel therapeutic strategies targeting mitochondrial biogenesis for T2DM treatment and its associated complications. It also discusses several natural products that exhibit beneficial effects on T2DM by promoting mitochondrial biogenesis.
Collapse
Affiliation(s)
- Wenwen Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiaoxue Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Kaiyi Lai
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Ying Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
3
|
Hettler SA, Picascia T, Pastene DO, Vajpayee S, Perciaccante R, Yard BA, Gretz N, Krämer BK. Hyperfiltration can be detected by transcutaneous assessment of glomerular filtration rate in diabetic obese mice. Am J Physiol Cell Physiol 2023; 325:C1558-C1566. [PMID: 37955125 DOI: 10.1152/ajpcell.00483.2022] [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: 10/31/2022] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023]
Abstract
We addressed if hyperfiltration can be assessed transcutaneously in male diabetic obese mice (BTBRob/ob) at 12 and 24 wk and how this relates to glomerular parameters indicative for hyperfiltration. Transcutaneous assessment of FITC-Sinistrin clearance [transcutaneous assessment of glomerular filtration rate (tGFR)] was compared against classical plasma clearance. Kidney from SV620C-01-PEI perfused mice were harvested at 24 wk and processed for tissue clearing and classical histology. Perfusion patterns of glomerular capillaries, glomerular size, and vasodilation of the afferent arterioles were assessed. Although at 12 wk FITC-Sinistrin half-life (t1/2) for both tGFR and plasma clearance suggested hyperfiltration, this was not significant anymore at 24 wk. In kidneys of diabetic mice the diameter of the afferent arteriole was significantly larger and positively correlated with glomerular size. Glomerular perfusion pattern in these mice was heterogeneous ranging from non- to well-perfused glomeruli. Nonperfused glomerular areas displayed a strong periodic acid-Schiff's (PAS) positive staining. Collectively our data demonstrate that tGFR is a valid method to detect hyperfiltration. Hyperfiltration occurs early in BTBRob/ob mice and disappears with disease progression as a consequence of a reduced filtration surface. It remains to be assessed if tGFR is also a valid method in diabetic mice with severely compromised renal function.NEW & NOTEWORTHY tGFR measurement is a relatively new method to assess kidney function in conscious rodents, which can be repeated multiple times in the same animal to track the course of the disease and/or the effect of potential treatments. Since the literature was inconclusive on the suitability of this technique in obese mice, we validated it for the first time against classical plasma clearance in the commonly used BTBRob/ob mouse model.
Collapse
Affiliation(s)
- Steffen A Hettler
- Vth Medical Department, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Tiziana Picascia
- Medical Research Center, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Diego O Pastene
- V Medical Department, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | - Benito A Yard
- V Medical Department, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Bernhard K Krämer
- V Medical Department, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
4
|
Wang F, Liu C, Ren L, Li Y, Yang H, Yu Y, Xu W. Sanziguben polysaccharides improve diabetic nephropathy in mice by regulating gut microbiota to inhibit the TLR4/NF-κB/NLRP3 signalling pathway. PHARMACEUTICAL BIOLOGY 2023; 61:427-436. [PMID: 36772833 PMCID: PMC9930838 DOI: 10.1080/13880209.2023.2174145] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 06/02/2023]
Abstract
CONTEXT Sanziguben (SZGB) is an empirical prescription used in traditional Chinese medicine to treat diabetic nephropathy (DN). As an abundant and primarily effective component of SZGB, Sanziguben polysaccharides (SZP) can be digested by flora to generate biological activity. OBJECTIVE Our study aimed to clarify the potential mechanism of SZP in improving chronic DN. MATERIALS AND METHODS Male db/db mice were randomized into DN, SZP (500 mg/kg) and metformin (MET, 300 mg/kg) groups. Wild-type littermates served as the normal control (NC) group. The drug was orally administered for 8 weeks. Enzyme-linked immunosorbent assay was used to detect the inflammatory factors. Proteins related to inflammation were evaluated using western blotting and immunohistochemical examination. Gut microbiota was analysed using 16S rRNA sequencing. RESULTS SZP significantly reduced 24 h urine albumin (p < 0.05) of DN mice. Compared to DN group, SZP significantly decreased the homeostasis model assessment of insulin resistance index, serum creatinine and blood urea nitrogen levels (20.27 ± 3.50 vs. 33.64 ± 4.85, 19.22 ± 3.77 vs. 32.52 ± 3.05 μmol/L, 13.23 ± 1.42 vs. 16.27 ± 0.77 mmol/L, respectively), and mitigated renal damage. SZP also regulated gut microbiota and decreased the abundance of Gram-negative bacteria (Proteobacteria, Klebsiella and Escherichia-Shigella). Subsequently, SZP reduced lipopolysaccharides levels (1.06- to 1.93-fold) of DN mice. Furthermore, SZP inhibited the expression levels of TLR4, phospho-NF-κB p65, NLRP3 proteins and interleukin (IL)-18 and IL-1β. CONCLUSIONS These results demonstrated that SZP improved intestinal flora disorder and inhibited the TLR4/NF-κB/NLRP3 pathway to alleviate DN.
Collapse
Affiliation(s)
- Fan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - LingZhi Ren
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - YanYang Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - HongMei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Yu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - WeiPing Xu
- Nuclear Medicine Department, Guangdong Provincial Peoples Hospital, Guangzhou, China
| |
Collapse
|
5
|
Yu Q, Chen Y, Zhao Y, Huang S, Xin X, Jiang L, Wang H, Wu W, Qu L, Xiang C, Wang S, Liu G, Yang L. Nephropathy Is Aggravated by Fatty Acids in Diabetic Kidney Disease through Tubular Epithelial Cell Necroptosis and Is Alleviated by an RIPK-1 Inhibitor. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:408-423. [PMID: 37927402 PMCID: PMC10624943 DOI: 10.1159/000529995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 11/07/2023]
Abstract
Introduction Diabetic kidney disease (DKD), one of the leading causes of end-stage renal disease, has complex pathogenic mechanisms and few effective clinical therapies. DKD progression is accompanied by the loss of renal resident cells, followed by chronic inflammation and extracellular matrix deposition. Necroptosis is a newly discovered form of regulated cell death and is a major form of intrinsic cell loss in certain diabetic complications such as cardiomyopathy, intestinal disease, and retinal neuropathy; however, its significance in DKD is largely unknown. Methods In this study, the expression of necroptosis marker phosphorylated MLKL (p-MLKL) in renal biopsy tissues of patients with DKD was detected using immunofluorescence and semiquantified using immunohistochemistry. The effects of different disease-causing factors on necroptosis activation in human HK-2 cells were evaluated using immunofluorescence and Western blotting. db/db diabetic mice were fed a high-fat diet to establish an animal model of DKD with significant renal tubule damage. Mice were treated with the RIPK1 inhibitor RIPA-56 to evaluate its renal protective effects. mRNA transcriptome sequencing was used to explore the changes in signaling pathways after RIPA-56 treatment. Oil red O staining and electron macroscopy were used to observe lipid droplet accumulation in renal biopsy tissues and mouse kidney tissues. Results Immunostaining of phosphorylated RIPK1/RIPK3/MLKL verified the occurrence of necroptosis in renal tubular epithelial cells of patients with DKD. The level of the necroptosis marker p-MLKL correlated positively with the severity of renal functional, pathological damages, and lipid droplet accumulation in patients with DKD. High glucose and fatty acids were the main factors causing necroptosis in human renal tubular HK-2 cells. Renal function deterioration and renal pathological injury were accelerated, and the necroptosis pathway was activated in db/db mice fed a high-fat diet. Application of RIPA-56 effectively reduced the degree of renal injury, inhibited the necroptosis pathway activation, and reduced necroinflammation and lipid droplet accumulation in the renal tissues of db/db mice fed a high-fat diet. Conclusion The present study revealed the role of necroptosis in the progression of DKD and might provide a new therapeutic target for the treatment of DKD.
Collapse
Affiliation(s)
- Qi Yu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Ying Chen
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Youlu Zhao
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuo Huang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Xiaohong Xin
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Lei Jiang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Wenyan Wu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Lei Qu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Chengang Xiang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Suxia Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Gang Liu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Ashfaq A, Meineck M, Pautz A, Arioglu-Inan E, Weinmann-Menke J, Michel MC. A systematic review on renal effects of SGLT2 inhibitors in rodent models of diabetic nephropathy. Pharmacol Ther 2023; 249:108503. [PMID: 37495021 DOI: 10.1016/j.pharmthera.2023.108503] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023]
Abstract
We have performed a systematic review of studies reporting on the renal effects of SGLT2 inhibitors in rodent models of diabetes. In 105 studies, SGLT2 inhibitors improved not only the glycemic control but also various aspects of renal function in most cases. These nephroprotective effects were similarly reported whether treatment with the SGLT2 inhibitor started concomitant with the onset of diabetes (within 1 week), early after onset (1-4 weeks) or after nephropathy had developed (>4 weeks after onset) with the latter probably having the greatest translational value. They were observed across various animal models of type 1 and type 2 diabetes/obesity (4 and 23 models, respectively), although studies in the type 2 diabetes model of db/db mice more often had negative data than in other models. Among possibly underlying pathophysiological mechanisms of nephroprotection, treatment with SGLT2 inhibitors had beneficial effects on lipid metabolism, blood pressure, glomerulosclerosis as well as renal tubular fibrosis, apoptosis, oxidative stress, and inflammation. These pathomechanisms highly influence atherosclerosis and renal health, which are two major factors that lead to an enhanced mortality in patients with diabetes and/or chronic kidney disease. Interestingly, renal SGLT2 inhibitor effects did not always correlate with those on glucose homeostasis, particularly in a limited number of direct comparative studies with other anti-diabetic treatments, indicating that nephroprotection may at least partly occur by mechanisms other than improving glycemic control. Our analyses did not provide evidence for different nephroprotective efficacy between SGLT2 inhibitors. Importantly, only four of 105 studies reported on female animals, and none provided direct comparative data between sexes. We conclude that more data on female animals and more direct comparative studies with other anti-diabetic compounds and combinations of treatments are needed.
Collapse
Affiliation(s)
- Aqsa Ashfaq
- Dept. of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Myriam Meineck
- 1(st) Dept. of Medicine, Div. of Nephrology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Andrea Pautz
- Dept. of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ebru Arioglu-Inan
- Dept. of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Julia Weinmann-Menke
- 1(st) Dept. of Medicine, Div. of Nephrology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Martin C Michel
- Dept. of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
7
|
Rakhat Y, Wang L, Han W, Rustemova A, Kulzhanova N, Yamada Y, Yabe D, Seino Y, Yada T. Oral Semaglutide under Human Protocols and Doses Regulates Food Intake, Body Weight, and Glycemia in Diet-Induced Obese Mice. Nutrients 2023; 15:3765. [PMID: 37686797 PMCID: PMC10489807 DOI: 10.3390/nu15173765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The first oral form of the glucagon-like peptide-1 receptor agonist, oral semaglutide, has recently been launched and potently controls glycemia and body weight in subjects with type 2 diabetes. This drug carries the absorption enhancer and requires specific protocols of administration. The mechanism of action of oral semaglutide is not fully understood, for which an appropriate experimental model is required. This study explores the metabolic effects of oral semaglutide in mice under human protocols and doses. Oral semaglutide was bolus and once daily injected into high-fat diet-induced obese (DIO) mice under human protocols, followed by monitoring blood glucose, food intake, and body weight. Oral semaglutide 0.23 mg/kg, a comparable human dose (14 mg) in a small volume of water under human protocols rapidly decreased blood glucose and food intake and continuously reduced food intake and weight gain for 3 days in DIO mice. At 0.7 mg/kg (42 mg), this drug was somewhat more potent. Oral semaglutide with human protocols and doses rapidly reduces blood glucose and food intake and continuously suppresses feeding and weight in DIO mice. This study establishes mice as a model suitable for analyzing the mechanism of anti-obesity/diabetes actions of oral semaglutide.
Collapse
Affiliation(s)
- Yermek Rakhat
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Lei Wang
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Wanxin Han
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Aktolkyn Rustemova
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Nazymgul Kulzhanova
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuichiro Yamada
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu 501-1193, Japan
| | - Yutaka Seino
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu 501-1193, Japan
| |
Collapse
|
8
|
Liu Z, Liu J, Wang W, An X, Luo L, Yu D, Sun W. Epigenetic modification in diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1133970. [PMID: 37455912 PMCID: PMC10348754 DOI: 10.3389/fendo.2023.1133970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
Diabetic kidney disease (DKD) is a common microangiopathy in diabetic patients and the main cause of death in diabetic patients. The main manifestations of DKD are proteinuria and decreased renal filtration capacity. The glomerular filtration rate and urinary albumin level are two of the most important hallmarks of the progression of DKD. The classical treatment of DKD is controlling blood glucose and blood pressure. However, the commonly used clinical therapeutic strategies and the existing biomarkers only partially slow the progression of DKD and roughly predict disease progression. Therefore, novel therapeutic methods, targets and biomarkers are urgently needed to meet clinical requirements. In recent years, increasing attention has been given to the role of epigenetic modification in the pathogenesis of DKD. Epigenetic variation mainly includes DNA methylation, histone modification and changes in the noncoding RNA expression profile, which are deeply involved in DKD-related inflammation, oxidative stress, hemodynamics, and the activation of abnormal signaling pathways. Since DKD is reversible at certain disease stages, it is valuable to identify abnormal epigenetic modifications as early diagnosis and treatment targets to prevent the progression of end-stage renal disease (ESRD). Because the current understanding of the epigenetic mechanism of DKD is not comprehensive, the purpose of this review is to summarize the role of epigenetic modification in the occurrence and development of DKD and evaluate the value of epigenetic therapies in DKD.
Collapse
Affiliation(s)
- Zhe Liu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Jiahui Liu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Wanning Wang
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingna An
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Luo
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Weixia Sun
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
9
|
Luo W, Tang S, Xiao X, Luo S, Yang Z, Huang W, Tang S. Translation Animal Models of Diabetic Kidney Disease: Biochemical and Histological Phenotypes, Advantages and Limitations. Diabetes Metab Syndr Obes 2023; 16:1297-1321. [PMID: 37179788 PMCID: PMC10168199 DOI: 10.2147/dmso.s408170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Animal models play a crucial role in studying the pathogenesis of diseases, developing new drugs, identifying disease risk markers, and improving means of prevention and treatment. However, modeling diabetic kidney disease (DKD) has posed a challenge for scientists. Although numerous models have been successfully developed, none of them can encompass all the key characteristics of human DKD. It is essential to choose the appropriate model according to the research needs, as different models develop different phenotypes and have their limitations. This paper provides a comprehensive overview of biochemical and histological phenotypes, modeling mechanisms, advantages and limitations of DKD animal models, in order to update relevant model information and provide insights and references for generating or selecting the appropriate animal models to fit different experimental needs.
Collapse
Affiliation(s)
- Wenting Luo
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Xiang Xiao
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Simin Luo
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Zixuan Yang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Songqi Tang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| |
Collapse
|
10
|
Kamli-Salino SEJ, Brown PAJ, Haschler TN, Liang L, Feliers D, Wilson HM, Delibegovic M. Induction of experimental diabetes and diabetic nephropathy using anomer-equilibrated streptozotocin in male C57Bl/6J mice. Biochem Biophys Res Commun 2023; 650:109-116. [PMID: 36774688 DOI: 10.1016/j.bbrc.2023.01.089] [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: 01/10/2023] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 02/08/2023]
Abstract
Streptozotocin (STZ) is widely used to induce experimental diabetes in murine models. However, the ability to induce diabetic nephropathy (DN) is more challenging. It has been recommended to inject STZ at multiple low doses within 15 min after dissolution due to its alleged instability. However, some studies suggest that STZ is stable for days due to equilibration of its two anomers (α and β), 90 min after dissolution, and that this anomer-equilibrated STZ leads to higher survival rates and persistent hyperglycaemia with minimal weight loss. The aim of this study was to determine an optimal dose of anomer-equilibrated STZ to induce kidney tubular damage and compare it with the more commonly used freshly prepared STZ. We hypothesised that anomer-equilibrated STZ provides a better, reproducible experimental model of diabetes-induced kidney damage with improved animal welfare. Body measurements, fasting glycaemia, insulinemia and renal histology were assessed in male C57Bl/6J at two and six months of age treated with fresh (50 mg/kg) or anomer-equilibrated (dose ranging 35-50 mg/kg) STZ or vehicle control. We demonstrated a dose-dependent effect of anomer-equilibrated STZ on the induction of hypo-insulinaemia and hyperglycaemia, as well as body weight in two-month-old mice. Interestingly, in six-month-old mice STZ leads to body weight loss, independently of STZ preparation mode. Anomer-equilibrated STZ provoked moderate to severe kidney tubule structural damage, resulting in significant kidney hypertrophy, whereas freshly prepared STZ only caused mild alterations. In conclusion, our study proposes that anomer-equilibrated STZ provides a robust murine model of diabetes and early-stage diabetic nephropathy, which can be used to test therapeutic approaches to treat and/or prevent renal damage.
Collapse
Affiliation(s)
- Sarah E J Kamli-Salino
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, UK.
| | - Paul A J Brown
- Department of Pathology, Aberdeen Royal Infirmary, Foresterhill, Aberdeen, Scotland, UK
| | - Timo N Haschler
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal & Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Lihuan Liang
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal & Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Denis Feliers
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal & Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Heather M Wilson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, UK
| | - Mirela Delibegovic
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
11
|
Tao Y, Young‐Stubbs C, Yazdizadeh Shotorbani P, Su D, Mathis KW, Ma R. Sex and strain differences in renal hemodynamics in mice. Physiol Rep 2023; 11:e15644. [PMID: 36946063 PMCID: PMC10031302 DOI: 10.14814/phy2.15644] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 03/23/2023] Open
Abstract
The present study was to examine sex and strain differences in glomerular filtration rate (GFR) and renal blood flow (RBF) in C57BL6, 129/Sv, and C57BLKS/J mice, three commonly used mouse strains in renal research. GFR was measured by transdermal measurement of FITC-sinitrin clearance in conscious mice. RBF was measured by a flow probe placed in the renal artery under an anesthetic state. In C57BL6 mice, there were no sex differences in both GFR and RBF. In 129/Sv mice, females had significantly greater GFR than males at age of 24 weeks, but not at 8 weeks. However, males had higher RBF and lower renal vascular resistance (RVR). Similar to 129/Sv, female C57BLKS/J had significantly greater GFR at both 8 and 24 weeks, lower RBF, and higher RVR than males. Across strains, male 129/Sv had lower GFR and higher RBF than male C57BL6, but no significant difference in GFR and greater RBF than male C57BLKS/J. No significant difference in GFR or RBF was observed between C57BL6 and C57BLKS/J mice. Deletion of eNOS in C57BLKS/J mice reduced GFR in both sexes, but decreased RBF in males. Furthermore, there were no sex differences in the severity of renal injury in eNOS-/- dbdb mice. Taken together, our study suggests that sex differences in renal hemodynamics in mice are strain and age dependent. eNOS was not involved in the sex differences in GFR, but in RBF. Furthermore, the sexual dimorphism did not impact the severity of renal injury in diabetic nephropathy.
Collapse
Affiliation(s)
- Yu Tao
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Cassandra Young‐Stubbs
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | | | - Dong‐Ming Su
- Department of Microbiology, Immunology and GeneticsUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Keisa W. Mathis
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Rong Ma
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
12
|
Mullins TP, Schock-Kusch D, Gallo LA. Transdermal Measurement of Glomerular Filtration Rate in Preclinical Research. Methods Mol Biol 2023; 2664:309-315. [PMID: 37423996 DOI: 10.1007/978-1-0716-3179-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The measurement of glomerular filtration rate (GFR) is essential to understanding renal physiology, including the monitoring of disease progression and treatment effectiveness. Transdermal measurement of glomerular filtration rate (tGFR) using a miniaturized fluorescence monitor in combination with a fluorescent exogenous GFR tracer has become a common technique to measure GFR in the preclinical setting, especially in rodent models. It allows for close to real-time measurement of GFR in conscious unrestrained animals and overcomes several limitations of other GFR measures. Its widespread use is reflected by published research articles and conference abstracts from different research fields, including in the assessment of new and existing kidney therapeutics, evaluation of nephrotoxicity, screening of novel chemical or medical agents, and fundamental understanding of kidney function.
Collapse
Affiliation(s)
- Thomas P Mullins
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Daniel Schock-Kusch
- Institute of Process Control and Innovative Energy Conversion, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Linda A Gallo
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, Australia.
| |
Collapse
|
13
|
Sun X, Wang G. Renal outcomes with sodium-glucose cotransporters 2 inhibitors. Front Endocrinol (Lausanne) 2022; 13:1063341. [PMID: 36531469 PMCID: PMC9752889 DOI: 10.3389/fendo.2022.1063341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the most serious complications of diabetes. Therefore, delaying and preventing the progression of DN becomes an important goal in the clinical treatment of type 2 diabetes mellitus. Recent studies confirm that sodium-glucose cotransporters 2 inhibitors (SGLT2is) have been regarded as effective glucose-lowering drugs with renal protective effect. In this review, we summarize in detail the present knowledge of the effects of SGLT2is on renal outcomes by analyzing the experimental data in preclinical study, the effects of SGLT2is on estimated glomerular flitration rates (eGFRs) and urinary albumin-creatinine ratios (UACRs) from clinical trials and observational studies, and renal events (such as renal death or renal failure requiring renal replacement therapy) in some large prospective cardiovaslucar outcomes trials. The underlying mechanisms for renoprotective activity of SGLT2is have been demondtrated in multiple diabetic and nondiabetic animal models including kidney-specific effects and secondary kidney effects related to amelioration in blood glucose and blood pressure. In conclusion, these promising results show that SGLT2is act beneficially in terms of the kidney for diabetic patients.
Collapse
Affiliation(s)
| | - Guohong Wang
- Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Vergara A, Jacobs-Cacha C, Llorens-Cebria C, Ortiz A, Martinez-Diaz I, Martos N, Dominguez-Báez P, Van den Bosch MM, Bermejo S, Pieper MP, Benito B, Soler MJ. Enhanced Cardiorenal Protective Effects of Combining SGLT2 Inhibition, Endothelin Receptor Antagonism and RAS Blockade in Type 2 Diabetic Mice. Int J Mol Sci 2022; 23:12823. [PMID: 36361612 PMCID: PMC9656616 DOI: 10.3390/ijms232112823] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 07/30/2023] Open
Abstract
Treatments with sodium-glucose 2 cotransporter inhibitors (SGLT2i) or endothelin receptor antagonists (ERA) have shown cardiorenal protective effects. The present study aimed to evaluate the cardiorenal beneficial effects of the combination of SGLT2i and ERA on top of renin-angiotensin system (RAS) blockade. Type 2 diabetic mice (db/db) were treated with different combinations of an SGLT2i (empagliflozin), an ERA (atrasentan), and an angiotensin-converting enzyme inhibitor (ramipril) for 8 weeks. Vehicle-treated diabetic mice and non-diabetic mice were included as controls. Weight, blood glucose, blood pressure, and kidney and heart function were monitored during the study. Kidneys and heart were collected for histological examination and to study the intrarenal RAS. Treatment with empagliflozin alone or combined significantly decreased blood glucose compared to vehicle-treated db/db. The dual and triple therapies achieved significantly greater reductions in diastolic blood pressure than ramipril alone. Compared to vehicle-treated db/db, empagliflozin combined with ramipril or in triple therapy significantly prevented GFR increase, but only the triple combination exerted greater protection against podocyte loss. In the heart, empagliflozin alone or combined reduced cardiac isovolumetric relaxation time (IVRT) and left atrium (LA) diameter as compared to vehicle-treated db/db. However, only the triple therapy was able to reduce cardiomyocyte area. Importantly, the add-on triple therapy further enhanced the intrarenal ACE2/Ang(1-7)/Mas protective arm of the RAS. These data suggest that triple therapy with empagliflozin, atrasentan and ramipril show synergistic cardiorenal protective effects in a type 2 diabetic mouse model.
Collapse
Affiliation(s)
- Ander Vergara
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Nephrology Department, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Conxita Jacobs-Cacha
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Carmen Llorens-Cebria
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Alberto Ortiz
- IIS-Fundación Jiménez Diaz, Fundación Renal Iñigo Álvarez de Toledo-IRSIN, REDinREN, Instituto de Investigación Carlos III, Universidad Autónoma de Madrid, Av. de los Reyes Católicos 2, 28040 Madrid, Spain
| | - Irene Martinez-Diaz
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Nerea Martos
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Pamela Dominguez-Báez
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Mireia Molina Van den Bosch
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Sheila Bermejo
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Nephrology Department, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Michael Paul Pieper
- Cardio-Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397 Biberach an der Riß, Germany
| | - Begoña Benito
- Cardiology Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Cardiology Department, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Deparment of Medicine, Universitat Autònoma de Barcelona, Av. de Can Domènech, 08193 Bellaterra, Spain
| | - Maria Jose Soler
- Nephrology and Kidney Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Nephrology Department, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
15
|
Efficacy of Dapagliflozin in Patients with Diabetes Mellitus Complicated with Coronary Artery Disease and Its Impact on the Vascular Endothelial Function. DISEASE MARKERS 2022; 2022:4829750. [PMID: 36118673 PMCID: PMC9481371 DOI: 10.1155/2022/4829750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022]
Abstract
Objective. To investigate the efficacy of dapagliflozin for diabetes mellitus complicated by coronary artery diseases and its impact on vascular endothelial function. Methods. Between August 2020 and August 2021, 80 patients with coronary heart disease complicated by type 2 diabetes mellitus were recruited and randomly assigned to receive either dapagliflozin (5 mg daily) plus original oral hypoglycemic agents (dapagliflozin group) or original oral hypoglycemic agents alone (control group). Outcome measures included blood pressure, blood glucose, cholesterol levels, vascular endothelial function, cardiovascular events, and drug-related adverse events. Results. The two groups had similar outcome indices upon admission (
). After 20 weeks of medication, the two groups of patients showed similar blood pressure, hemoglobin A1c (HbA1c), and low-density lipoprotein (LDL-C) levels versus those before treatment (
), and no significant differences were found in intergroup comparison neither (
). Dapagliflozin plus conventional hypoglycemic agents resulted in a significantly higher reactive hyperemia index (RHI) value, fewer cases with abnormal vascular endothelial function, and fewer major cardiovascular events during treatment versus the sole use of conventional hypoglycemic agents (
). There was no significant difference in drug-related adverse events between the two groups (
). Conclusion. Dapagliflozin improves the vascular endothelial functions of patients with diabetes mellitus complicated by coronary artery disease with a high safety profile and favorable efficacy.
Collapse
|
16
|
Palacios-Ramirez R, Lima-Posada I, Bonnard B, Genty M, Fernandez-Celis A, Hartleib-Geschwindner J, Foufelle F, Lopez-Andres N, Bamberg K, Jaisser F. Mineralocorticoid Receptor Antagonism Prevents the Synergistic Effect of Metabolic Challenge and Chronic Kidney Disease on Renal Fibrosis and Inflammation in Mice. Front Physiol 2022; 13:859812. [PMID: 35464084 PMCID: PMC9022039 DOI: 10.3389/fphys.2022.859812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/21/2022] [Indexed: 01/09/2023] Open
Abstract
Obesity and/or metabolic diseases are frequently associated with chronic kidney disease and several factors associated with obesity may contribute to proteinuria and extracellular matrix production. Mineralocorticoid receptor antagonists have proven their clinical efficacy in diabetic kidney disease with preclinical data suggesting that they may also be efficient in non-diabetic chronic kidney disease associated to metabolic diseases. In the present study we developed a novel mouse model combining severe nephron reduction and High Fat Diet challenge that led to chronic kidney disease with metabolic alterations. We showed that the Mineralocorticoid Receptor antagonist canrenoate improved metabolic function, reduced albuminuria and prevented the synergistic effect of high fat diet on renal fibrosis and inflammation in chronic kidney disease mice.
Collapse
Affiliation(s)
- Roberto Palacios-Ramirez
- Centre de Recherche des Cordeliers, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Ixchel Lima-Posada
- Centre de Recherche des Cordeliers, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Benjamin Bonnard
- Centre de Recherche des Cordeliers, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Marie Genty
- Centre de Recherche des Cordeliers, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Amaya Fernandez-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Judith Hartleib-Geschwindner
- Research and Early Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Fabienne Foufelle
- Centre de Recherche des Cordeliers, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Natalia Lopez-Andres
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Krister Bamberg
- Research and Early Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Frederic Jaisser
- Centre de Recherche des Cordeliers, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, Inserm, Université de Paris, Paris, France
- Université de Lorraine, INSERM Centre D’Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France
- *Correspondence: Frederic Jaisser,
| |
Collapse
|
17
|
Microencapsulated islet transplantation alleviates podocyte injury in diabetic nephropathy via inhibiting Notch-1 signaling. Transpl Immunol 2022; 72:101579. [PMID: 35278650 DOI: 10.1016/j.trim.2022.101579] [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: 01/21/2022] [Revised: 03/06/2022] [Accepted: 03/06/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Podocyte injury has a critical role in the pathogenesis of diabetic nephropathy (DN). Microencapsulated islet transplantation (MIT) is identified as an effective method for improving the clinical condition of DN. This study aimed to explore the role and mechanism of MIT in alleviating podocyte injury in DN. METHODS A mouse model of DN was constructed using streptozotocin (STZ). Mice were divided into 3 groups: the untreated diabetic nephropathy group (DN group), the microencapsulated islet transplantation-treated group (MIT group) and the control group. The mice were raised for 6 weeks posterior to islet transplantation to identify the role of MIT. Renal function and structure of glomerular filtration barrier were assessed by urine analysis, histopathological examination, and transmission electron microscopy. The expression levels of several proteins including Caspase-3, Bcl2/Bax, β-galactosidase, Ki-67, synaptopodin, WT-1, Jagged-1, Notch-1, and Hes-1 in renal tissues were identified via immunohistochemistry (IHC), immunofluorescence (IF), and western blotting techniques. RESULTS Compared with the DN group, the MIT group presented decreased levels of blood glucose, urinary albumin/creatinine, urea nitrogen, and serum creatinine while their body weight gradually increased. Glomerular injury in the MIT group was significantly better than that in the DN group. The MIT group indicated significantly decreased expression of Caspase-3, β-galactosidase, Bax/Bcl-2, and Ki-67 when compared with DN group, while the proportion of synaptopodin- and WT-1-positive cells was significantly increased (P < 0.05). The protein expression of Jagged-1, Notch-1, and Hes-1 in the glomerulus of the MIT group was significantly lower than that in the DN group (P < 0.05). CONCLUSION MIT alleviates podocyte injury induced by DN by inhibiting Notch-1 signaling. The identification of signaling pathways influencing podocyte restoration can help evaluate personalized medicine efficacy for patients treated with islet transplantation.
Collapse
|
18
|
Revisiting glomerular hyperfiltration and examining the concept of high dietary protein-related nephropathy in athletes and bodybuilders. Curr Opin Nephrol Hypertens 2022; 31:18-25. [PMID: 34846311 DOI: 10.1097/mnh.0000000000000755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW High-protein diets (HPDs) are popular but their consequences for kidney health, especially among athletes and bodybuilders who typically maintain a high protein intake for a long time, have not been investigated. This review focused on recent studies of the association of HPD with long-term kidney health and the concept of high dietary protein-related nephropathy. RECENT FINDINGS Several long-term observational studies including large populations have reinforced the notion that HPDs are associated with a rapid decline of kidney function. An increase in renal blood flow and glomerular hyperfiltration caused by vasodilation, and increased levels of endocrine and paracrine factors (glucagon, IGF-1, prostanoids, and nitric oxide), facilitates the excretion of protein-derived nitrogenous waste. Inhibition of tubule-glomerular feedback and increased proximal tubular Na+ reabsorption after a HPD augment glomerular hyperfiltration and may trigger synthesis of proinflammatory cytokines and receptor for advanced glycation end-products (RAGE). Focal segmental glomerulosclerosis reported in association with anabolic steroid may indeed be a HPD nephropathy given that HPD results in progressive glomerulosclerosis, especially in remnant glomeruli or in diabetic kidney disease but can happen in any high-risk situation, such as solitary kidney and polycystic kidneys. SUMMARY HPD among athletes and bodybuilders in an extreme way across a long-term period may pose a risk to renal health including high incidence of HPD nephropathy.
Collapse
|
19
|
Østergaard MV, Secher T, Christensen M, Salinas CG, Roostalu U, Skytte JL, Rune I, Hansen HH, Jelsing J, Vrang N, Fink LN. Therapeutic effects of lisinopril and empagliflozin in a mouse model of hypertension-accelerated diabetic kidney disease. Am J Physiol Renal Physiol 2021; 321:F149-F161. [PMID: 34180715 DOI: 10.1152/ajprenal.00154.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hypertension is a critical comorbidity for progression of diabetic kidney disease (DKD). To facilitate the development of novel therapeutic interventions with the potential to control disease progression, there is a need to establish translational animal models that predict treatment effects in human DKD. The present study aimed to characterize renal disease and outcomes of standard of medical care in a model of advanced DKD facilitated by adeno-associated virus (AAV)-mediated renin overexpression in uninephrectomized (UNx) db/db mice. Five weeks after single AAV administration and 4 wk after UNx, female db/db UNx-ReninAAV mice received (PO, QD) vehicle, lisinopril (40 mg/kg), empagliflozin (20 mg/kg), or combination treatment for 12 wk (n = 17 mice/group). Untreated db/+ mice (n = 8) and vehicle-dosed db/db UNx-LacZAAV mice (n = 17) served as controls. End points included plasma, urine, and histomorphometric markers of kidney disease. Total glomerular numbers and individual glomerular volume were evaluated by whole kidney three-dimensional imaging analysis. db/db UNx-ReninAAV mice developed hallmarks of progressive DKD characterized by severe albuminuria, advanced glomerulosclerosis, and glomerular hypertrophy. Lisinopril significantly improved albuminuria, glomerulosclerosis, tubulointerstitial injury, and inflammation. Although empagliflozin alone had no therapeutic effect on renal endpoints, lisinopril and empagliflozin exerted synergistic effects on renal histological outcomes. In conclusion, the db/db UNx-ReninAAV mouse demonstrates good clinical translatability with respect to physiological and histological hallmarks of progressive DKD. The efficacy of standard of care to control hypertension and hyperglycemia provides a proof of concept for testing novel drug therapies in the model.NEW & NOTEWORTHY Translational animal models of diabetic kidney disease (DKD) are important tools in preclinical research and drug discovery. Here, we show that the standard of care to control hypertension (lisinopril) and hyperglycemia (empagliflozin) improves physiological and histopathological hallmarks of kidney disease in a mouse model of hypertension-accelerated progressive DKD. The findings substantiate hypertension and type 2 diabetes as essential factors in driving DKD progression and provide a proof of concept for probing novel drugs for potential nephroprotective efficacy in this model.
Collapse
|
20
|
Renal hemodynamic effects of sodium-glucose cotransporter 2 inhibitors in hyperfiltering people with type 1 diabetes and people with type 2 diabetes and normal kidney function. Kidney Int 2021; 97:631-635. [PMID: 32200854 DOI: 10.1016/j.kint.2019.12.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 01/15/2023]
|
21
|
Knopp T, Bieler T, Jung R, Ringen J, Molitor M, Jurda A, Münzel T, Waisman A, Wenzel P, Karbach SH, Wild J. Effects of Dietary Protein Intake on Cutaneous and Systemic Inflammation in Mice with Acute Experimental Psoriasis. Nutrients 2021; 13:nu13061897. [PMID: 34072973 PMCID: PMC8228490 DOI: 10.3390/nu13061897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Psoriasis is a systemic inflammatory disorder, primarily characterized by skin plaques. It is linked to co-morbidities including cardiovascular disease and metabolic syndrome. Several studies demonstrate that dietary habits can influence psoriasis development and severity. However, the effect of different dietary protein levels on psoriasis development and severity is poorly understood. In this study, we examine the influence of dietary protein on psoriasis-like skin disease in mice. Methods: We fed male C57BL/6J mice with regular, low protein and high protein chow for 4 weeks. Afterwards, we induced psoriasis-like skin disease by topical imiquimod (IMQ)-treatment on ear and back skin. The local cutaneous and systemic inflammatory response was investigated using flow cytometry analysis, histology and quantitative rt-PCR. Results: After 5 days of IMQ-treatment, both diets reduced bodyweight in mice, whereas only the high protein diet slightly aggravated IMQ-induced skin inflammation. IMQ-treatment induced infiltration of myeloid cells, neutrophils, and monocytes/macrophages into skin and spleen independently of diet. After IMQ-treatment, circulating neutrophils and reactive oxygen species were increased in mice on low and high protein diets. Conclusion: Different dietary protein levels had no striking effect on IMQ-induced psoriasis but aggravated the systemic pro-inflammatory phenotype.
Collapse
Affiliation(s)
- Tanja Knopp
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
| | - Tabea Bieler
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
| | - Rebecca Jung
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
| | - Julia Ringen
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
| | - Michael Molitor
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
- Center for Cardiology—Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany;
- German Center for Cardiovascular Research (DZHK)—Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Annika Jurda
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
| | - Thomas Münzel
- Center for Cardiology—Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany;
- German Center for Cardiovascular Research (DZHK)—Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Ari Waisman
- Institute of Molecular Medicine, University Medical Center Mainz, 55131 Mainz, Germany;
- Focus Program Translational Neurosciences, University Medical Center Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
- Center for Cardiology—Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany;
- German Center for Cardiovascular Research (DZHK)—Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Susanne Helena Karbach
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
- Center for Cardiology—Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany;
- German Center for Cardiovascular Research (DZHK)—Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Johannes Wild
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany; (T.K.); (T.B.); (R.J.); (J.R.); (M.M.); (A.J.); (P.W.); (S.H.K.)
- Center for Cardiology—Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany;
- German Center for Cardiovascular Research (DZHK)—Partner Site Rhine-Main, 55131 Mainz, Germany
- Correspondence:
| |
Collapse
|
22
|
Snelson M, Clarke RE, Nguyen TV, Penfold SA, Forbes JM, Tan SM, Coughlan MT. Long Term High Protein Diet Feeding Alters the Microbiome and Increases Intestinal Permeability, Systemic Inflammation and Kidney Injury in Mice. Mol Nutr Food Res 2021; 65:e2000851. [PMID: 33547877 DOI: 10.1002/mnfr.202000851] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/07/2021] [Indexed: 02/06/2023]
Abstract
SCOPE This study evaluates the effects of a chronic high protein diet (HPD) on kidney injury, intestinal permeability and gut microbiota perturbations in a mouse model. METHOD AND RESULTS Mice are fed a diet containing either 20% or 52% energy from protein for 24 weeks; protein displaced an equivalent amount of wheat starch. The HPD does not alter glycemic control or body weight. The HPD induces kidney injury as evidenced by increase in albuminuria, urinary kidney injury molecule-1, blood urea nitrogen, urinary isoprostanes and renal cortical NF-κB p65 gene expression. HPD decreases intestinal occludin gene expression, increases plasma endotoxin and plasma monocyte chemoattractant protein-1, indicating intestinal leakiness and systemic inflammation. Cecal microbial analysis reveals that HPD feeding does not alter alpha diversity; however, it does alter beta diversity, indicating an altered microbial community structure with HPD feeding. Predicted metagenome pathway analysis demonstrates a reduction in branched-chain amino acid synthesis and an increase of the urea cycle with consumption of a HPD. CONCLUSION These results demonstrate that long term HPD consumption in mice causes albuminuria, systemic inflammation, increase in gastrointestinal permeability and is associated with gut microbiome remodeling with an increase in the urea cycle pathway, which may contribute to renal injury.
Collapse
Affiliation(s)
- Matthew Snelson
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Rachel Elise Clarke
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | | | - Sally Anne Penfold
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Josephine Maree Forbes
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Sih Min Tan
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Melinda Therese Coughlan
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
23
|
Sembach FE, Østergaard MV, Vrang N, Feldt-Rasmussen B, Fosgerau K, Jelsing J, Fink LN. Rodent models of diabetic kidney disease: human translatability and preclinical validity. Drug Discov Today 2021; 26:200-217. [DOI: 10.1016/j.drudis.2020.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
|
24
|
Mazzucato M, Fioretto P, Avogaro A. High-protein diet: A barrier to the nephroprotective effects of sodium-glucose co-transporter-2 inhibitors? Diabetes Obes Metab 2020; 22:1511-1515. [PMID: 32350981 DOI: 10.1111/dom.14071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022]
Abstract
Glomerular hyperfiltration is a common finding in patients with diabetes and poor glycaemic control; whole-kidney hyperfiltration, with glomerular filtration rate (GFR) values above normal, should be differentiated from single nephron hyperfiltration, consequent to nephron loss and compensatory hyperfiltration of the remnant nephrons. This is the result of an imbalance between the vascular tone of the afferent and efferent arterioles. Hormonal influences and/or an impaired tubuloglomerular feedback (TGF) system, because of excessive sodium (Na+ ) and glucose reabsorption in the proximal tubule, contribute to determine hyperfiltration. Sodium-glucose co-transporter-2 inhibitors (SGLT2is), by decreasing Na+ reabsorption and increasing the delivery of Na+ to the macula densa, lead to normalization of TGF, and, consequently, decrease GFR (both whole and single nephron). High-protein diets are popular among patients with type 1 and type 2 diabetes; importantly, 80% of the amino acids are also reabsorbed in the proximal tubule of the nephron and are transported by symporters that use the electro-chemical gradient of Na+ . Indeed, an acute protein load is associated with increased Na+ reabsorption and an increase in GFR. Here, we hypothesize that high-protein diets, by increasing Na+ reabsorption and GFR, may offset the positive renal effects of SGLT2is.
Collapse
Affiliation(s)
| | - Paola Fioretto
- Department of Medicine, University of Padova, Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
25
|
Zhan M, Kanwar YS. An enigma: does a high-protein diet accelerate renal damage in humans? Lessons from diabetic animal models. Am J Physiol Renal Physiol 2020; 318:F979-F981. [PMID: 32174145 DOI: 10.1152/ajprenal.00076.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Ming Zhan
- Department of Nephrology, Ningbo First Hospital, Zhejiang University, China.,Department of Pathology and Medicine, Northwestern University, Chicago, Illinois
| | - Yashpal S Kanwar
- Department of Pathology and Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
26
|
Packer M. Autophagy stimulation and intracellular sodium reduction as mediators of the cardioprotective effect of sodium-glucose cotransporter 2 inhibitors. Eur J Heart Fail 2020; 22:618-628. [PMID: 32037659 DOI: 10.1002/ejhf.1732] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022] Open
Abstract
In five large-scale trials involving >40 000 patients, sodium-glucose cotransporter 2 (SGLT2) inhibitors decreased the risk of serious heart failure events by 25-40%. This effect cannot be explained by control of hyperglycaemia, since it is not observed with antidiabetic drugs with greater glucose-lowering effects. It cannot be attributed to ketogenesis, since it is not causally linked to ketone body production, and the benefit is not enhanced in patients with diabetes. The effect cannot be ascribed to a natriuretic action, since SGLT2 inhibitors decrease natriuretic peptides only modestly, and they reduce cardiovascular death, a benefit that diuretics do not possess. Although SGLT2 inhibitors increase red blood cell mass, enhanced erythropoiesis does not favourably influence the course of heart failure. By contrast, experimental studies suggest that SGLT2 inhibitors may reduce intracellular sodium, thereby preventing oxidative stress and cardiomyocyte death. Additionally, SGLT2 inhibitors induce a transcriptional paradigm that mimics nutrient and oxygen deprivation, which includes activation of adenosine monophosphate-activated protein kinase, sirtuin-1, and/or hypoxia-inducible factors-1α/2α. The interplay of these mediators stimulates autophagy, a lysosomally-mediated degradative pathway that maintains cellular homeostasis. Autophagy-mediated clearance of damaged organelles reduces inflammasome activation, thus mitigating cardiomyocyte dysfunction and coronary microvascular injury. Interestingly, the action of hypoxia-inducible factors-1α/2α to both stimulate erythropoietin and induce autophagy may explain why erythrocytosis is strongly correlated with the reduction in heart failure events. Therefore, the benefits of SGLT2 inhibitors on heart failure may be mediated by a direct cardioprotective action related to modulation of pathways responsible for cardiomyocyte homeostasis.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX, USA.,Imperial College, London, UK
| |
Collapse
|
27
|
Bessho R, Takiyama Y, Takiyama T, Kitsunai H, Takeda Y, Sakagami H, Ota T. Hypoxia-inducible factor-1α is the therapeutic target of the SGLT2 inhibitor for diabetic nephropathy. Sci Rep 2019; 9:14754. [PMID: 31611596 PMCID: PMC6791873 DOI: 10.1038/s41598-019-51343-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
Previous studies have demonstrated intrarenal hypoxia in patients with diabetes. Hypoxia-inducible factor (HIF)-1 plays an important role in hypoxia-induced tubulointerstitial fibrosis. Recent clinical trials have confirmed the renoprotective action of SGLT2 inhibitors in diabetic nephropathy. We explored the effects of an SGLT2 inhibitor, luseogliflozin on HIF-1α expression in human renal proximal tubular epithelial cells (HRPTECs). Luseogliflozin significantly inhibited hypoxia-induced HIF-1α protein expression in HRPTECs. In addition, luseogliflozin inhibited hypoxia-induced the expression of the HIF-1α target genes PAI-1, VEGF, GLUT1, HK2 and PKM. Although luseogliflozin increased phosphorylated-AMP-activated protein kinase α (p-AMPKα) levels, the AMPK activator AICAR did not changed hypoxia-induced HIF-1α expression. Luseogliflozin suppressed the oxygen consumption rate in HRPTECs, and subsequently decreased hypoxia-sensitive dye, pimonidazole staining under hypoxia, suggesting that luseogliflozin promoted the degradation of HIF-1α protein by redistribution of intracellular oxygen. To confirm the inhibitory effect of luseogliflozin on hypoxia-induced HIF-1α protein in vivo, we treated male diabetic db/db mice with luseogliflozin for 8 to 16 weeks. Luseogliflozin attenuated cortical tubular HIF-1α expression, tubular injury and interstitial fibronectin in db/db mice. Together, luseogliflozin inhibits hypoxia-induced HIF-1α accumulation by suppressing mitochondrial oxygen consumption. The SGLT2 inhibitors may protect diabetic kidneys by therapeutically targeting HIF-1α protein.
Collapse
Affiliation(s)
- Ryoichi Bessho
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Yumi Takiyama
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan.
| | - Takao Takiyama
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hiroya Kitsunai
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Yasutaka Takeda
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hidemitsu Sakagami
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Tsuguhito Ota
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|