1
|
Ryytty S, Nurminen K, Mäkinen P, Suomalainen A, Hämäläinen RH. Heightened sensitivity to adverse effects of metformin in mtDNA mutant patient cells. Life Sci 2025; 366-367:123486. [PMID: 39978587 DOI: 10.1016/j.lfs.2025.123486] [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: 11/04/2024] [Revised: 01/23/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
AIMS Metformin (Met) is a widely used, cost-effective, and relatively safe drug, primarily prescribed for diabetes, that also exhibits beneficial effects in other conditions, such as in cardiovascular diseases, neurological disorders, and cancer. Despite its common use, the safety of Met in patients with primary mitochondrial disease remains uncertain, as both Met and mitochondrial dysfunction increase the risk of lactic acidosis. Here we have examined the effects of Met in patient cells with m.3243A>G mitochondrial DNA mutation. MATERIALS AND METHODS We utilized induced pluripotent stem cells (iPSCs) derived from two m.3243A>G patients, alongside cardiomyocytes differentiated from these iPSCs (iPSC-CMs). The cells were exposed to 10, 100, and 1000 μM Met for 24 h, and the effects on cellular metabolism and mitochondrial function were evaluated. KEY FINDINGS While low concentrations, relative to common therapeutic plasma levels, increased mitochondrial respiration, higher concentrations decreased respiration in both patient and control cells. Furthermore, cells with high level of the m.3243A>G mutation were more sensitive to Met than control cells. Additionally, we observed a clear patient-specific response to Met in cardiomyocytes. SIGNIFICANCE The findings emphasize the critical importance of selecting appropriate Met concentrations in cellular experiments and demonstrate the variability in Met's effects between individuals. Moreover, the results highlight the need for caution when considering Met use in patients with primary mitochondrial disorders.
Collapse
Affiliation(s)
- Sanna Ryytty
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Katriina Nurminen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Anu Suomalainen
- Stem Cell and Metabolism Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland; HUSLab, Helsinki University Hospital, Helsinki, Finland
| | | |
Collapse
|
2
|
Gojani EG, Wang B, Li D, Kovalchuk O, Kovalchuk I. Single and Combined Impact of Semaglutide, Tirzepatide, and Metformin on β-Cell Maintenance and Function Under High-Glucose-High-Lipid Conditions: A Comparative Study. Int J Mol Sci 2025; 26:421. [PMID: 39796271 PMCID: PMC11720205 DOI: 10.3390/ijms26010421] [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: 12/02/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/30/2025] Open
Abstract
Type 2 diabetes (T2D), the most common form, is marked by insulin resistance and β-cell failure. β-cell dysfunction under high-glucose-high-lipid (HG-HL) conditions is a key contributor to the progression of T2D. This study evaluates the comparative effects of 10 nM semaglutide, 10 nM tirzepatide, and 1 mM metformin, both alone and in combination, on INS-1 β-cell maintenance and function under HG-HL conditions. INS-1 cells were pretreated for 2 h with single doses of metformin (1 mM), semaglutide (10 nM), tirzepatide (10 nM), or combinations of 1 mM metformin with either 10 nM semaglutide or 10 nM tirzepatide, followed by 48 h of HG-HL stimulation. The results indicate that combining 1 mM metformin with either 10 nM semaglutide or 10 nM tirzepatide significantly enhances the effects of 10 nM semaglutide and 10 nM tirzepatide on HG-HL-induced apoptosis and dysregulated cell cycle. Specifically, the combination treatments demonstrated superior restoration of glucose-stimulated insulin secretion (GSIS) functionality compared to 1 mM metformin, 10 nM semaglutide, and 10 nM tirzepatide.
Collapse
Affiliation(s)
| | | | | | | | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.L.); (O.K.)
| |
Collapse
|
3
|
Miguel V, Shaw IW, Kramann R. Metabolism at the crossroads of inflammation and fibrosis in chronic kidney disease. Nat Rev Nephrol 2025; 21:39-56. [PMID: 39289568 DOI: 10.1038/s41581-024-00889-z] [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: 08/15/2024] [Indexed: 09/19/2024]
Abstract
Chronic kidney disease (CKD), defined as persistent (>3 months) kidney functional loss, has a growing prevalence (>10% worldwide population) and limited treatment options. Fibrosis driven by the aberrant accumulation of extracellular matrix is the final common pathway of nearly all types of chronic repetitive injury in the kidney and is considered a hallmark of CKD. Myofibroblasts are key extracellular matrix-producing cells that are activated by crosstalk between damaged tubules and immune cells. Emerging evidence indicates that metabolic alterations are crucial contributors to the pathogenesis of kidney fibrosis by affecting cellular bioenergetics and metabolite signalling. Immune cell functions are intricately connected to their metabolic characteristics, and kidney cells seem to undergo cell-type-specific metabolic shifts in response to damage, all of which can determine injury and repair responses in CKD. A detailed understanding of the heterogeneity in metabolic reprogramming of different kidney cellular subsets is essential to elucidating communication processes between cell types and to enabling the development of metabolism-based innovative therapeutic strategies against CKD.
Collapse
Affiliation(s)
- Verónica Miguel
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Isaac W Shaw
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Rafael Kramann
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany.
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Zhu J, Li L, Luan Y, Zhang Z, Wang Y, Xu Z. Salidroside Pre-Treatment Inhibits Hypertensive Renal Injury and Fibrosis Through Inhibiting Wnt/β-Catenin Pathway. Dose Response 2024; 22:15593258241298045. [PMID: 39506979 PMCID: PMC11539081 DOI: 10.1177/15593258241298045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Objectives This study aimed to explore the protective effects and underlying mechanisms of salidroside (SAL) in angiotensin II (Ang II)-induced hypertensive renal injury and fibrosis, using in vivo and in vitro models. Methods In this study, we generated Ang II-induced hypertensive renal injury and fibrosis in mice and the recombinant interferon-gamma (IFN-γ)-stimulated murine podocyte clone 5 (MPC5) model in vitro. Histological and oxidative stress analyses were performed to evaluate the renal injury. Results SAL pre-treatment reduced systolic blood pressure (SBP), diastolic blood pressure (DBP), mean arterial blood pressure (MAP), and attenuated serum creatinine (Scr), blood urea nitrogen (BUN), and serum cystatin C (Cys-C) levels in Ang II-infused mice (all, P < 0.001). SAL reduced renal fibrosis and related molecules expression, including Collagen I, Collagen III, and α-smooth muscle actin (α-SMA) (all, P < 0.001). SAL decreased the content of malondialdehyde (MDA) while increasing superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) in Ang II-treated mice (all, P < 0.001). In addition, SAL pre-treatment inhibited AT1R, Wnt1, Wnt3a, and β-catenin expressions (all, P < 0.001), both in vivo and in vitro. Conclusion Our experimental data demonstrate that SAL pre-treatment protects against Ang II-induced hypertensive renal injury and fibrosis by suppressing the Wnt/β-catenin pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Emergency Medicine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Li
- Department of Emergency Medicine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuting Luan
- Department of Infectious Diseases, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziqing Zhang
- Department of Emergency Medicine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Wang
- Department of Emergency Medicine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenyu Xu
- Department of Emergency Medicine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Zheng Q, Zhao J, Yuan J, Qin Y, Zhu Z, Liu J, Sun S. Delaying Renal Aging: Metformin Holds Promise as a Potential Treatment. Aging Dis 2024:AD.2024.0168. [PMID: 39012670 DOI: 10.14336/ad.2024.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/06/2024] [Indexed: 07/17/2024] Open
Abstract
Given the rapid aging of the population, age-related diseases have become an excessive burden on global health care. The kidney, a crucial metabolic organ, ages relatively quickly. While the aging process itself does not directly cause kidney damage, the physiological changes that accompany it can impair the kidney's capacity for self-repair. This makes aging kidneys more susceptible to diseases, including increased risks of chronic kidney disease and end-stage renal disease. Therefore, delaying the progression of renal aging and preserving the youthful vitality of the kidney are crucial for preventing kidney diseases. However, effective strategies against renal aging are still lacking due to the underlying mechanisms of renal aging, which have not been fully elucidated. Accumulating evidence suggests that metformin has beneficial effects in mitigating renal aging. Metformin has shown promising anti-aging results in animal models but has not been tested for this purpose yet in clinical trials. These findings indicate the potential of metformin as an anti-renal aging drug. In this review, we primarily discuss the characteristics and mechanisms of kidney aging and the potential effects of metformin against renal aging.
Collapse
Affiliation(s)
- Qiao Zheng
- Department of Postgraduate Student, Xi'an Medical University, Xi'an, China
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jin Zhao
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinguo Yuan
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunlong Qin
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhanxin Zhu
- Department of Postgraduate Student, Xi'an Medical University, Xi'an, China
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Zang L, Saitoh S, Katayama K, Zhou W, Nishimura N, Shimada Y. A zebrafish model of diabetic nephropathy shows hyperglycemia, proteinuria and activation of the PI3K/Akt pathway. Dis Model Mech 2024; 17:dmm050438. [PMID: 38747698 DOI: 10.1242/dmm.050438] [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: 08/03/2023] [Accepted: 05/08/2024] [Indexed: 05/30/2024] Open
Abstract
Diabetic nephropathy (DN), as a complication of diabetes, is a substantial healthcare challenge owing to the high risk of morbidity and mortality involved. Although significant progress has been made in understanding the pathogenesis of DN, more efficient models are required to develop new therapeutics. Here, we created a DN model in zebrafish by crossing diabetic Tg(acta1:dnIGF1R-EGFP) and proteinuria-tracing Tg(l-fabp::VDBP-GFP) lines, named zMIR/VDBP. Overfed adult zMIR/VDBP fish developed severe hyperglycemia and proteinuria, which were not observed in wild-type zebrafish. Renal histopathology revealed human DN-like characteristics, such as glomerular basement membrane thickening, foot process effacement and glomerular sclerosis. Glomerular dysfunction was restored upon calorie restriction. RNA sequencing analysis demonstrated that DN zebrafish kidneys exhibited transcriptional patterns similar to those seen in human DN pathogenesis. Notably, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway was activated, a phenomenon observed in the early phase of human DN. In addition, metformin improved hyperglycemia and proteinuria in DN zebrafish by modulating Akt phosphorylation. Our results indicate that zMIR/VDBP fish are suitable for elucidating the mechanisms underlying human DN and could be a powerful tool for therapeutic discovery.
Collapse
Affiliation(s)
- Liqing Zang
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Mie 514-8507, Japan
- Mie University Zebrafish Research Center, Tsu, Mie 514-8507, Japan
| | - Sei Saitoh
- Department of Biomedical Molecular Sciences (Anatomy II), Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Kan Katayama
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Weibin Zhou
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY 10029-5674, USA
| | - Norihiro Nishimura
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Mie 514-8507, Japan
- Mie University Zebrafish Research Center, Tsu, Mie 514-8507, Japan
| | - Yasuhito Shimada
- Mie University Zebrafish Research Center, Tsu, Mie 514-8507, Japan
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
7
|
Kleibert M, Zygmunciak P, Łakomska K, Mila K, Zgliczyński W, Mrozikiewicz-Rakowska B. Insight into the Molecular Mechanism of Diabetic Kidney Disease and the Role of Metformin in Its Pathogenesis. Int J Mol Sci 2023; 24:13038. [PMID: 37685845 PMCID: PMC10487922 DOI: 10.3390/ijms241713038] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 09/10/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the leading causes of death among patients diagnosed with diabetes mellitus. Despite the growing knowledge about the pathogenesis of DKD, we still do not have effective direct pharmacotherapy. Accurate blood sugar control is essential in slowing down DKD. It seems that metformin has a positive impact on kidneys and this effect is not only mediated by its hypoglycemic action, but also by direct molecular regulation of pathways involved in DKD. The molecular mechanism of DKD is complex and we can distinguish polyol, hexosamine, PKC, and AGE pathways which play key roles in the development and progression of this disease. Each of these pathways is overactivated in a hyperglycemic environment and it seems that most of them may be regulated by metformin. In this article, we summarize the knowledge about DKD pathogenesis and the potential mechanism of the nephroprotective effect of metformin. Additionally, we describe the impact of metformin on glomerular endothelial cells and podocytes, which are harmed in DKD.
Collapse
Affiliation(s)
- Marcin Kleibert
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Przemysław Zygmunciak
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (P.Z.); (K.M.)
| | - Klaudia Łakomska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Klaudia Mila
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (P.Z.); (K.M.)
| | - Wojciech Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland;
| | - Beata Mrozikiewicz-Rakowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland;
| |
Collapse
|
8
|
Peng W, Yang B, Qiao H, Liu Y, Lin Y. Metformin use is associated with reduced acute kidney injury following coronary artery bypass grafting in patients with type 2 diabetes: An inverse probability of treatment weighting analysis. Pharmacotherapy 2023; 43:778-786. [PMID: 37199291 DOI: 10.1002/phar.2827] [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: 02/18/2023] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 05/19/2023]
Abstract
STUDY OBJECTIVE Acute kidney injury (AKI) is a common and serious complication after coronary artery bypass grafting (CABG) surgery. Patients with diabetes are commonly associated with renal microvascular complications and have a greater risk of AKI after CABG surgery. This study aimed to explore whether preoperative metformin administration could reduce the incidence of postoperative AKI following CABG in patients with type 2 diabetes. DESIGN Patients with diabetes who underwent CABG were retrospectively included in this study. AKI after CABG was defined according to the Kidney Disease Improving Global Outcomes (KDIGO) criteria. The effects of metformin on postoperative AKI following CABG in patients were compared and analyzed. DATA SOURCE Patients were enrolled in this study between January 2019 and December 2020 in Beijing Anzhen Hospital. PATIENTS A total of 812 patients were enrolled. The patients were divided into the metformin group (203 cases) and the control group (609 cases) according to whether metformin was used preoperatively. INTERVENTION Inverse probability of treatment weighting (IPTW) was applied to minimize baseline differences between the two groups. IPT-weighted p values were analyzed to evaluate the postoperative outcomes between the two groups. MEASUREMENTS AND MAIN RESULTS The incidence of AKI in the metformin group and the control group was compared. After IPTW adjustment, the incidence of AKI in the metformin group was lower than the control group (IPTW-adjusted p < 0.001). In the subgroup analysis, metformin showed significant protective effects in the estimated glomerular filtration rate (eGFR) < 60 mL/min per 1.73 m2 and eGFR 60-90 mL/min per 1.73 m2 subgroups, which was not observed in the eGFR ≥90 mL/min per 1.73 m2 subgroup. No significant differences in the incidence of renal replacement therapy, reoperation due to bleeding, in-hospital mortality, or red blood cell transfusion volume were observed between the two groups. CONCLUSIONS In this study, we provided evidence that preoperative metformin was associated with a significant reduction of postoperative AKI following CABG in patients with diabetes. Metformin showed significant protective effects in patients with mild-to-moderate renal insufficiency.
Collapse
Affiliation(s)
- Wenxing Peng
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bo Yang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huanyu Qiao
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yongmin Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Lin
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Ahmed AA, Mohamed SK, Nofal S, El Morsy EM, Ahmed AAE. Effect of bempedoic acid on angiotensin-II induced hypertension and vascular tissue remodelling in renal hypertensive rats through AMPK multiple signalling pathways modulation. Life Sci 2023; 320:121573. [PMID: 36931497 DOI: 10.1016/j.lfs.2023.121573] [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: 11/23/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Angiotensin II (Ang II), the effector of the renin-angiotensin system (RAS), is a key player in the pathogenesis of chronic hypertension, accompanied by vascular tissue resistance, remodelling, and damage. Chronic activation of Ang II receptor 1 (AT-1R) impairs multiple cellular targets implicated in cellular protection and survival, including adenosine Monophosphate-activated protein kinase (AMPK) signalling. In addition, it induces oxidative damage, endoplasmic reticulum (ER) stress, and fibrotic changes in resistance vessels. Our study investigated the antihypertensive and antifibrotic effects of bempedoic acid, a first-in-class antihyperlipidemic drug that targets adenosine triphosphate-citrate lyase enzyme to inhibit cholesterol synthesis. We also studied the modulation of multiple AMPK signalling pathways by bempedoic acid in a chronic hypertension model in rats. Sixty male Sprague-Dawley rats were divided into four groups: sham group, hypertensive group, standard captopril group, and bempedoic treated group. All groups underwent left renal artery ligation except the sham group. Fourteen days post-surgery, captopril and bempedoic acid were administered with a dose of 30 mg/kg/day orally to captopril-standard and bempedoic acid-treated groups for two weeks, respectively. In mesenteric resistance arteries, bempedoic acid activated AMPK energy independently and augmented AMPK multiple cellular targets to adapt to Ang II-induced cellular stress. It exerted antioxidant activity, increased endothelial nitric oxide synthase, and reversed the ER stress. Bempedoic acid maintained vascular integrity and prevented vascular remodelling by inhibiting extracellular signal-regulated kinase (ERK)/transforming growth factor-β fibrotic pathway. These effects were reflected in the improved hemodynamic measurements.
Collapse
Affiliation(s)
- Asmaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt.
| | - Shimaa K Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| | - Engy M El Morsy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| |
Collapse
|
10
|
Pavlovic K, Krako Jakovljevic N, Isakovic AM, Ivanovic T, Markovic I, Lalic NM. Therapeutic vs. Suprapharmacological Metformin Concentrations: Different Effects on Energy Metabolism and Mitochondrial Function in Skeletal Muscle Cells in vitro. Front Pharmacol 2022; 13:930308. [PMID: 35873556 PMCID: PMC9299382 DOI: 10.3389/fphar.2022.930308] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022] Open
Abstract
Metformin is an oral antidiabetic agent that has been widely used in clinical practice for over 60 years, and is currently the most prescribed antidiabetic drug worldwide. However, the molecular mechanisms of metformin action in different tissues are still not completely understood. Although metformin-induced inhibition of mitochondrial respiratory chain Complex I and activation of AMP-activated protein kinase have been observed in many studies, published data is inconsistent. Furthermore, metformin concentrations used for in vitro studies and their pharmacological relevance are a common point of debate. The aim of this study was to explore the effects of different metformin concentrations on energy metabolism and activity of relevant signaling pathways in C2C12 muscle cells in vitro. In order to determine if therapeutic metformin concentrations have an effect on skeletal muscle cells, we used micromolar metformin concentrations (50 µM), and compared the effects with those of higher, millimolar concentrations (5 mM), that have already been established to affect mitochondrial function and AMPK activity. We conducted all experiments in conditions of high (25 mM) and low glucose (5.5 mM) concentration, in order to discern the role of glucose availability on metformin action. According to our results, micromolar metformin treatment did not cause Complex I inhibition nor AMPK activation. Also, cells cultured in low glucose medium were more sensitive to Complex I inhibition, mitochondrial membrane depolarization and AMPK activation by millimolar metformin, but cells cultured in high glucose medium were more prone to induction of ROS production. In conclusion, even though suprapharmacological metformin concentrations cause Complex I inhibition and AMPK activation in skeletal muscle cells in vitro, therapeutic concentrations cause no such effect. This raises the question if these mechanisms are relevant for therapeutic effects of metformin in skeletal muscle.
Collapse
Affiliation(s)
- Kasja Pavlovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Nina Krako Jakovljevic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Andjelka M Isakovic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Tijana Ivanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivanka Markovic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Nebojsa M Lalic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
11
|
Rogacka D, Piwkowska A. Beneficial effects of metformin on glomerular podocytes in diabetes. Biochem Pharmacol 2021; 192:114687. [PMID: 34274355 DOI: 10.1016/j.bcp.2021.114687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 01/15/2023]
Abstract
Podocytes and their foot processes form an important cellular layer of the glomerular barrier involved in regulating glomerular permeability. Disturbances in podocyte function play a central role in the development of proteinuria in diabetic nephropathy. The retraction of podocyte foot processes forming a slit diaphragm is a common feature of proteinuria. Metformin is an oral antidiabetic agent of the biguanide class that is widely recommended for the treatment of high blood glucose in patients with type 2 diabetes mellitus. In addition to lowering glucose, several recent studies have reported potential beneficial effects of metformin on diabetic kidney function. Furthermore, a key molecule of the antidiabetic mechanism of action of metformin is adenosine 5'-monophospate-activated protein kinase (AMPK), as the metformin-induced activation of AMPK is well documented. The present review summarizes current knowledge on the protective effects of metformin against pathological changes in podocytes that are induced by hyperglycemia.
Collapse
Affiliation(s)
- Dorota Rogacka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences, Wita Stwosza 63, Gdansk 80-308, Poland; Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences, Wita Stwosza 63, Gdansk 80-308, Poland; Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland.
| |
Collapse
|
12
|
Shi Z, Chen G, Cao Z, Wu F, Lei H, Chen C, Song Y, Liu C, Li J, Zhou J, Lu Y, Zhang L. Gut Microbiota and Its Metabolite Deoxycholic Acid Contribute to Sucralose Consumption-Induced Nonalcoholic Fatty Liver Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3982-3991. [PMID: 33755449 DOI: 10.1021/acs.jafc.0c07467] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
As important signal metabolites within enterohepatic circulation, bile acids (BAs) play a pivotal role during the occurrence and development of diet-induced nonalcoholic fatty liver disease (NAFLD). Here, we evaluated the functional effects of BAs and gut microbiota contributing to sucralose consumption-induced NAFLD of mice. The results showed that sucralose consumption significantly upregulated the abundance of intestinal genera Bacteroides and Clostridium, which produced deoxycholic acid (DCA) accumulating in multiple biological matrixes including feces, serum, and liver of mice. Subsequently, elevated hepatic DCA, one of the endogenous antagonists of the farnesol X receptor (Fxr), inhibited hepatic gene expression including a small heterodimer partner (Shp) and Fxr leading to sucralose-induced NAFLD in mice. Dietary supplements with fructo-oligosaccharide or metformin markedly restored genera Bacteroides and Clostridium abundance and the DCA level of sucralose-consuming mice, which eventually ameliorated NAFLD. These findings highlighted the effects of gut microbiota and its metabolite DCA on sucralose-induced NAFLD of mice.
Collapse
Affiliation(s)
- Zunji Shi
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
| | - Gui Chen
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng Cao
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Wu
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hehua Lei
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
| | - Chuan Chen
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuchen Song
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Caixiang Liu
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
| | - Jinquan Li
- Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinlin Zhou
- Engineering Research Academy of High Value Utilization of Green Plants, Meizhou 514021, China
| | - Yujing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
- Engineering Research Academy of High Value Utilization of Green Plants, Meizhou 514021, China
| | - Limin Zhang
- Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- Engineering Research Academy of High Value Utilization of Green Plants, Meizhou 514021, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
13
|
Song A, Zhang C, Meng X. Mechanism and application of metformin in kidney diseases: An update. Biomed Pharmacother 2021; 138:111454. [PMID: 33714781 DOI: 10.1016/j.biopha.2021.111454] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 01/08/2023] Open
Abstract
Metformin is an oral antihyperglycemic drug widely used to treat type 2 diabetes mellitus (T2DM), acting via indirect activation of 5' Adenosine monophosphate-activated Protein Kinase (AMPK). Beyond the anti-diabetic effect, accumulative pieces of evidence have revealed that metformin also everts a beneficial effect in diverse kidney diseases. In various acute kidney diseases (AKI) animal models, metformin protects renal tubular cells from inflammation, apoptosis, reactive oxygen stress (ROS), endoplasmic reticulum (ER) stress, epithelial-mesenchymal transition (EMT) via AMPK activation. In diabetic kidney disease (DKD), metformin also alleviates podocyte loss, mesangial cells apoptosis, and tubular cells senescence through AMPK-mediated signaling pathways. Besides, metformin inhibits cystic fibrosis transmembrane conductance regulator (CFTR)-mediated fluids secretion and the mammalian target of rapamycin (mTOR)-involved cyst formation negatively regulated by AMPK in autosomal dominant polycystic kidney disease (APDKD). Furthermore, metformin also contributes to the alleviation of urolithiasis and renal cell carcinoma (RCC). As the common pathway for chronic kidney disease (CKD) progressing towards end-stage renal disease (ESRD), renal fibrosis is ameliorated by metformin, to a great extent dependent on AMPK activation. However, clinical data are not always consistent with preclinical data, some clinical investigations showed the unmeaningful even detrimental effect of metformin on T2DM patients with kidney diseases. Most importantly, metformin-associated lactic acidosis (MALA) is a vital issue restricting the application of metformin. Thus, we conclude the application of metformin in kidney diseases and uncover the underlying molecular mechanisms in this review.
Collapse
Affiliation(s)
- Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xianfang Meng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
14
|
Giaccari A, Solini A, Frontoni S, Del Prato S. Metformin Benefits: Another Example for Alternative Energy Substrate Mechanism? Diabetes Care 2021; 44:647-654. [PMID: 33608326 PMCID: PMC7896249 DOI: 10.2337/dc20-1964] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/03/2020] [Indexed: 02/03/2023]
Abstract
Since the UK Prospective Diabetes Study (UKPDS), metformin has been considered the first-line medication for patients with newly diagnosed type 2 diabetes. Though direct evidence from specific trials is still lacking, several studies have suggested that metformin may protect from diabetes- and nondiabetes-related comorbidities, including cardiovascular, renal, neurological, and neoplastic diseases. In the past few decades, several mechanisms of action have been proposed to explain metformin's protective effects, none being final. It is certain, however, that metformin increases lactate production, concentration, and, possibly, oxidation. Once considered a mere waste product of exercising skeletal muscle or anaerobiosis, lactate is now known to act as a major energy shuttle, redistributed from production sites to where it is needed. Through the direct uptake and oxidation of lactate produced elsewhere, all end organs can be rapidly supplied with fundamental energy, skipping glycolysis and its possible byproducts. Increased lactate production (and consequent oxidation) could therefore be considered a positive mechanism of action of metformin, except when, under specific circumstances, metformin and lactate become excessive, increasing the risk of lactic acidosis. We are proposing that, rather than considering metformin-induced lactate production as dangerous, it could be considered a mechanism through which metformin exerts its possible protective effect on the heart, kidneys, and brain and, to some extent, its antineoplastic action.
Collapse
Affiliation(s)
- Andrea Giaccari
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Simona Frontoni
- Unit of Endocrinology, Diabetes and Metabolism, San Giovanni Calibita Fatebenefratelli Hospital, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Stefano Del Prato
- Section of Metabolic Diseases and Diabetes, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
15
|
Mariano F, Biancone L. Metformin, chronic nephropathy and lactic acidosis: a multi-faceted issue for the nephrologist. J Nephrol 2020; 34:1127-1135. [PMID: 33373028 PMCID: PMC8357762 DOI: 10.1007/s40620-020-00941-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022]
Abstract
Metformin is currently considered a first-line therapy in type 2 diabetic patients. After issuing warnings for decades about the risks of lactic acidosis in patients with chronic nephropathy, metformin is now being re-evaluated. The most recent evidence from the literature has demonstrated both a low, acceptable risk of lactic acidosis and a series of favorable effects, which go beyond its hypoglycemic activity. Patients treated with metformin show a significant mortality reduction and lower progression towards end-stage renal disease in comparison with those treated with other hypoglycemic drugs. Concerning lactic acidosis, in the last few years it has been shown how lactic acidosis almost always developed when patients kept taking the drug in the face of a concomitant disease or situation such as sepsis, fever, diarrhea, vomiting, which reduced metformin renal clearance. Actually, clearance of metformin is mainly renal, both by glomerular filtration and tubular secretion (apparent clearance 933–1317 ml/min, half-life < 3 h). As regards treatment, in cases of lactic acidosis complicated by acute kidney injury, continuous renal replacement therapy (CRRT) plays a crucial role. Besides the elimination of metformin, CRRT improves survival by correcting acidosis, electrolyte alterations, and maintaining fluid balance. Lactic acidosis almost always develops because of preventable drug accumulation. Therefore, prevention is a key factor. Patients should be aware that discontinuation for a limited time does not affect their health, even when it may be inappropriate, but it may avoid a serious, potentially fatal adverse event.
Collapse
Affiliation(s)
- Filippo Mariano
- Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Luigi Biancone
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
16
|
Li L, Xie W, Gui Y, Zheng XL. Bromodomain-containing protein 4 and its role in cardiovascular diseases. J Cell Physiol 2020; 236:4829-4840. [PMID: 33345363 DOI: 10.1002/jcp.30225] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022]
Abstract
Bromodomain-containing protein 4 (BRD4), a chromatin-binding protein, is involved in the development of various tumors. Recent evidence suggests that BRD4 also plays a significant role in cardiovascular diseases, such as ischemic heart disease, hypertension, and cardiac hypertrophy. This review summarizes the roles of BRD4 as a potential regulator of various pathophysiological processes in cardiovascular diseases, implicating that BRD4 may be a new therapeutic target for cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Liang Li
- Department of Pathophysiology, Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Wei Xie
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada.,Department of Anatomy, Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, Hunan, China
| | - Yu Gui
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
17
|
Lehtonen S. Metformin Protects against Podocyte Injury in Diabetic Kidney Disease. Pharmaceuticals (Basel) 2020; 13:ph13120452. [PMID: 33321755 PMCID: PMC7764076 DOI: 10.3390/ph13120452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Metformin is the most commonly prescribed drug for treating type 2 diabetes mellitus (T2D). Its mechanisms of action have been under extensive investigation, revealing that it has multiple cellular targets, either direct or indirect ones, via which it regulates numerous cellular pathways. Diabetic kidney disease (DKD), the serious complication of T2D, develops in up to 50% of the individuals with T2D. Various mechanisms contribute to the development of DKD, including hyperglycaemia, dyslipidemia, oxidative stress, chronic low-grade inflammation, altered autophagic activity and insulin resistance, among others. Metformin has been shown to affect these pathways, and thus, it could slow down or prevent the progression of DKD. Despite several animal studies demonstrating the renoprotective effects of metformin, there is no concrete evidence in clinical settings. This review summarizes the renoprotective effects of metformin in experimental settings. Special emphasis is on the effects of metformin on podocytes, the glomerular epithelial cells that are central in maintaining the glomerular ultrafiltration function.
Collapse
Affiliation(s)
- Sanna Lehtonen
- Research Program for Clinical and Molecular Metabolism and Department of Pathology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
18
|
Rogacka D, Audzeyenka I, Piwkowska A. Regulation of podocytes function by AMP-activated protein kinase. Arch Biochem Biophys 2020; 692:108541. [PMID: 32781053 DOI: 10.1016/j.abb.2020.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/23/2020] [Accepted: 08/06/2020] [Indexed: 01/08/2023]
Abstract
Podocytes are unique, highly specialized, terminally differentiated cells that form an essential, integral part of the glomerular filter. These cells limit the outside border of the glomerular basement membrane, forming a tight barrier that prevents significant protein loss from the capillary space. The slit diaphragm formed by podocytes is crucial for maintaining glomerular integrity and function. They are the target of injury in many glomerular diseases, including hypertension and diabetes mellitus. Accumulating studies have revealed that AMP-activated protein kinase (AMPK), an essential cellular energy sensor, might play a fundamental role in regulating podocyte metabolism and function. AMPK participates in insulin signaling, therefore controls glucose uptake and podocytes insulin sensitivity. It is also involved in insulin-dependent cytoskeleton reorganization in podocytes, mediating glomerular albumin permeability. AMPK plays an important role in the regulation of autophagy/apoptosis processes, which influence podocytes viability. The present review aimed to highlight the molecular mechanisms associated with AMPK that are involved in the regulation of podocyte function in health and disease states.
Collapse
Affiliation(s)
- Dorota Rogacka
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| | - Irena Audzeyenka
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| |
Collapse
|
19
|
Eisenreich A, Orphal M, Böhme K, Kreutz R. Tmem63c is a potential pro-survival factor in angiotensin II-treated human podocytes. Life Sci 2020; 258:118175. [PMID: 32750436 DOI: 10.1016/j.lfs.2020.118175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/25/2020] [Accepted: 07/27/2020] [Indexed: 01/20/2023]
Abstract
AIMS Human podocytes (hPC) play an important role in the pathogenesis of renal diseases. In this context, angiotensin II (Ang II) and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) play a crucial role in podocyte injury. Recently, transmembrane protein (Tmem) 63c, a member of the Tmem-family was found to be expressed in kidney and associated with podocyte function. In this study, we analysed the expression regulation and functional impact of Tmem63c on cell viability and apoptosis in hPC in the context of Ang II activation. MATERIALS AND METHODS Expression of Tmem63c in response to Ang II and the NFκB inhibitor Bay 11-7082 was analysed by Real-Time PCR and Western blotting. Cellular functions were determined by functional assays. KEY FINDINGS We found Ang II to induce Tmem63c expression in hPC in a concentration-dependent manner. Inhibition of NFκB by Bay 11-7082 reduced basal as well as Ang II-induced Tmem63c expression. SiRNA-mediated down-regulation of Tmem63c diminished cell viability and protein kinase B (Akt) signaling and increased cell apoptosis of resting as well as Ang II-activated hPC. SIGNIFICANCE These data show that Ang II induced the expression of Tmem63c in hPC, possibly via NFκB-dependent mechanisms. Moreover, down-regulation of Tmem63c was associated with reduced cell viability, indicating Tmem63c to be a potential pro-survival factor in hPC.
Collapse
Affiliation(s)
- Andreas Eisenreich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Berlin, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany.
| | - Miriam Orphal
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Berlin, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Karen Böhme
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Berlin, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, 10115 Berlin, Germany
| | - Reinhold Kreutz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health Berlin, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, 10115 Berlin, Germany
| |
Collapse
|
20
|
Guo J, Zheng HJ, Zhang W, Lou W, Xia C, Han XT, Huang WJ, Zhang F, Wang Y, Liu WJ. Accelerated Kidney Aging in Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1234059. [PMID: 32774664 PMCID: PMC7407029 DOI: 10.1155/2020/1234059] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/25/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
With aging, the kidney undergoes inexorable and progressive changes in structural and functional performance. These aging-related alterations are more obvious and serious in diabetes mellitus (DM). Renal accelerated aging under DM conditions is associated with multiple stresses such as accumulation of advanced glycation end products (AGEs), hypertension, oxidative stress, and inflammation. The main hallmarks of cellular senescence in diabetic kidneys include cyclin-dependent kinase inhibitors, telomere shortening, and diabetic nephropathy-associated secretory phenotype. Lysosome-dependent autophagy and antiaging proteins Klotho and Sirt1 play a fundamental role in the accelerated aging of kidneys in DM, among which the autophagy-lysosome system is the convergent mechanism of the multiple antiaging pathways involved in renal aging under DM conditions. Metformin and the inhibitor of sodium-glucose cotransporter 2 are recommended due to their antiaging effects independent of antihyperglycemia, besides angiotensin-converting enzyme inhibitors/angiotensin receptor blockers. Additionally, diet intervention including low protein and low AGEs with antioxidants are suggested for patients with diabetic nephropathy (DN). However, their long-term benefits still need further study. Exploring the interactive relationships among antiaging protein Klotho, Sirt1, and autophagy-lysosome system may provide insight into better satisfying the urgent medical needs of elderly patients with aging-related DN.
Collapse
Affiliation(s)
- Jing Guo
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hui Juan Zheng
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wenting Zhang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wenjiao Lou
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chenhui Xia
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xue Ting Han
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jun Huang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Fan Zhang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yaoxian Wang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jing Liu
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Nephrology, and Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, No. 57th South Renmin Road, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
21
|
Metformin rescues Parkin protein expression and mitophagy in high glucose-challenged human renal epithelial cells by inhibiting NF-κB via PP2A activation. Life Sci 2020; 246:117382. [PMID: 32004509 DOI: 10.1016/j.lfs.2020.117382] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 12/20/2022]
Abstract
Our preliminary research revealed that metformin, a classic anti-diabetic drug, could rescue Parkin protein expression and mitophagy in high glucose-challenged human renal epithelial cells in vitro, but the molecular mechanism remains to be explored. In the study, Human Renal Cortical Epithelial Cells (HRCEpiC) and Human Renal Proximal Tubular Epithelial Cells (HRPTEpic) were challenged with high glucose with or without metformin pre-treatment to monitor Parkin mRNA and protein expression level change. PRKN gene knockdown was performed by lentiviral-based shRNA delivery. Cell viability, apoptosis and mitophagy were monitored after treatment. Mitochondrial damage was evaluated by analyzing mitochondrial permeability transition pore opening, membrane potential change, mitochondrial superoxide accumulation and cytochrome C release. Protein levels of activating transcription factor 4 (ATF4), p53 phospho-Ser15, IκBα phosphor-Ser32, IKKα phosphor-Ser176/180 in whole cell lysate and nuclear entry of p50/p65 were assessed by western blot. Okadaic acid was used to inhibit protein phosphatase 2A (PP2A). The data suggested high glucose challenge significantly reduced PRKN gene expression, mitophagy, mitochondria integrity and cell viability in vitro, which was rescued by metformin co-treatment. The effects of metformin were crippled by PRKN gene knockdown. Metformin increased PRKN gene transcription while reducednuclear factor kappa B (NF-κB) activation but not that of p53 or ATF4. Inhibiting PP2A weakened NF-κB inhibition and PRKN induction by metformin in high glucose-challenged cells, reducing its mitochondrial protective and cytoprotective effect. So, we concluded thatmetformin protects human renal epithelial cells from high glucose-induced apoptosis by restoring Parkin protein expression and mitophagy via PP2A activation and NF-κB inhibition.
Collapse
|
22
|
Akhter MS, Uppal P. Toxicity of Metformin and Hypoglycemic Therapies. Adv Chronic Kidney Dis 2020; 27:18-30. [PMID: 32146997 DOI: 10.1053/j.ackd.2019.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
Abstract
Metformin along with other antidiabetic medications provide benefit to patients in the treatment of type 2 diabetes mellitus, but caution is advised in certain scenarios to avoid toxicity in kidney disease. Renal dosing, monitoring of kidney function, and evaluating the risk of developing serious side effects are warranted with some agents. The available literature with regard to incidence of adverse events and toxicity of hypoglycemic therapies is reviewed.
Collapse
|
23
|
Lehtonen S. SHIPping out diabetes-Metformin, an old friend among new SHIP2 inhibitors. Acta Physiol (Oxf) 2020; 228:e13349. [PMID: 31342643 PMCID: PMC6916339 DOI: 10.1111/apha.13349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
SHIP2 (Src homology 2 domain‐containing inositol 5′‐phosphatase 2) belongs to the family of 5′‐phosphatases. It regulates the phosphoinositide 3‐kinase (PI3K)‐mediated insulin signalling cascade by dephosphorylating the 5′‐position of PtdIns(3,4,5)P3 to generate PtdIns(3,4)P2, suppressing the activity of the pathway. SHIP2 mouse models and genetic studies in human propose that increased expression or activity of SHIP2 contributes to the pathogenesis of the metabolic syndrome, hypertension and type 2 diabetes. This has raised great interest to identify SHIP2 inhibitors that could be used to design new treatments for metabolic diseases. This review summarizes the central mechanisms associated with the development of diabetic kidney disease, including the role of insulin resistance, and then moves on to describe the function of SHIP2 as a regulator of metabolism in mouse models. Finally, the identification of SHIP2 inhibitors and their effects on metabolic processes in vitro and in vivo are outlined. One of the newly identified SHIP2 inhibitors is metformin, the first‐line medication prescribed to patients with type 2 diabetes, further boosting the attraction of SHIP2 as a treatment target to ameliorate metabolic disorders.
Collapse
Affiliation(s)
- Sanna Lehtonen
- Department of Pathology and Research Program for Clinical and Molecular Metabolism, Faculty of Medicine University of Helsinki Helsinki Finland
| |
Collapse
|
24
|
Morresi C, Cianfruglia L, Sartini D, Cecati M, Fumarola S, Emanuelli M, Armeni T, Ferretti G, Bacchetti T. Effect of High Glucose-Induced Oxidative Stress on Paraoxonase 2 Expression and Activity in Caco-2 Cells. Cells 2019; 8:1616. [PMID: 31835890 PMCID: PMC6953021 DOI: 10.3390/cells8121616] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/14/2023] Open
Abstract
(1) Background: Hyperglycemia leads to several biochemical and physiological consequences, such as the generation of advanced glycation end products (AGEs) and reactive oxygen species (ROS), which are involved in the development of several human diseases. Intestinal cells are continuously exposed to pro-oxidants and lipid peroxidation products from ingested foods, and also to glyco-oxidative damage. It has been reported that free radical generation may be linked to the development of inflammation-related gastrointestinal diseases. (2) Methods: The effects of high glucose (HG) treatment (50 mM) were assessed in terms of free radical production, lipid peroxidation, and AGEs formation. Furthermore, the expression and the antiapoptotic and antioxidant activity of the paraoxonase-2 (PON2) enzyme in intestinal cells has been investigated. (3) Results: Caco-2 cells treated with media supplied with high glucose (HG) (50 mM) showed, with respect to physiological glucose concentration (25 mM), an increase in ROS production, lipid peroxidation, and AGEs formation. Moreover, a lower PON2 expression and activity in HG-treated cells was related to activation of the apoptotic pathways. (4) Conclusions: Our results demonstrated that high glucose concentrations triggered glyco-oxidative stress in intestinal cells; the downregulation of PON2 could result in a higher oxidative stress and might contribute to intestinal dysfunction.
Collapse
Affiliation(s)
- Camilla Morresi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (C.M.); (M.C.); (T.B.)
| | - Laura Cianfruglia
- Department of Clinical Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (L.C.); (D.S.); (S.F.); (M.E.)
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (L.C.); (D.S.); (S.F.); (M.E.)
| | - Monia Cecati
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (C.M.); (M.C.); (T.B.)
| | - Stefania Fumarola
- Department of Clinical Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (L.C.); (D.S.); (S.F.); (M.E.)
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (L.C.); (D.S.); (S.F.); (M.E.)
| | - Tatiana Armeni
- Department of Clinical Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (L.C.); (D.S.); (S.F.); (M.E.)
| | - Gianna Ferretti
- Department of Clinical Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (L.C.); (D.S.); (S.F.); (M.E.)
| | - Tiziana Bacchetti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (C.M.); (M.C.); (T.B.)
| |
Collapse
|
25
|
Lee MC, Lee CH, Chang LY, Chang CH, Zhang JF, Lee MR, Wang JY, Chen SM. Association of Metformin Use With End-Stage Renal Disease in Patients With Type 2 Diabetes Mellitus: A Nationwide Cohort Study Under the Pay-for-Performance Program. J Clin Pharmacol 2019; 59:1443-1452. [PMID: 31163098 DOI: 10.1002/jcph.1452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/16/2019] [Indexed: 12/25/2022]
Abstract
Animal studies have demonstrated that metformin exerts a renoprotective effect. Human studies of patients with diabetes mellitus (DM) regarding the association of metformin use with end-stage renal disease (ESRD) are lacking. Patients with type 2 DM and without a history of kidney disease who were enrolled under the pay-for-performance program of the National Health Insurance in Taiwan were identified. Those who received ≥90 cumulative defined daily doses of metformin within 1 year were selected (metformin users) and compared with a 1:1 propensity score-matched metformin nonuser cohort. Primary and secondary outcomes were development of ESRD and chronic kidney disease (CKD), respectively. Independent predictors were investigated using Cox regression analysis. A total of 24 158 pairs of metformin users and nonusers were enrolled, with an incidence of ESRD of 1908 and 1723 and CKD of 1095 and 1056 cases per 100 000 person-years, respectively. Metformin use was independently associated with increased risks of ESRD (adjusted hazard ratio, 1.22; 95% confidence interval, 1.12-1.32) and CKD (adjusted hazard ratio, 1.25; 95% confidence interval, 1.12-1.40) in a dose-response relationship. Patients with hypertension plus nonuse of angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers potentiated kidney damage by metformin. In patients with DM, use of metformin may increase the risk of ESRD and CKD. Health care professionals should be alert and closely monitor renal function when prescribing metformin.
Collapse
Affiliation(s)
- Ming-Chia Lee
- Department of Pharmacy, New Taipei City Hospital, New Taipei City, Taiwan.,School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsin Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lih-Yu Chang
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Chia-Hao Chang
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Jun-Fu Zhang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Meng-Rui Lee
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jann-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Ming Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
26
|
Lee J, Hong EM, Kim JH, Jung JH, Park SW, Koh DH, Choi MH, Jang HJ, Kae SH. Metformin Induces Apoptosis and Inhibits Proliferation through the AMP-Activated Protein Kinase and Insulin-like Growth Factor 1 Receptor Pathways in the Bile Duct Cancer Cells. J Cancer 2019; 10:1734-1744. [PMID: 31205529 PMCID: PMC6547996 DOI: 10.7150/jca.26380] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 01/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background/Aims: Metformin has been found to have antineoplastic activity in some cancer cells. This study was performed to determine whether metformin inhibits the proliferation of bile duct cancer cells by inducing apoptosis and its effects on the expression of gene-related proteins involved in cancer growth. Methods: Human extrahepatic bile duct cancer cells (SNU-245 and SNU-1196) were cultured. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays were performed to determine the effect of metformin on the cell proliferation. Apoptosis was measured by a cell death detection enzyme-linked immunosorbent assay and a caspase-3 activity assay. Expression levels of various proteins, with or without specific small interfering ribonucleic acid-induced gene disruption, were measured by Western blot analysis. The migratory activity of the cancer cells was evaluated by wound healing assay. Results: Metformin suppressed cell proliferation in bile duct cancer cells by inducing apoptosis. Metformin inhibited mammalian target of rapamycin (mTOR) by activation of tuberous sclerosis complex 2 (TSC-2) through phosphorylation of adenosine monophosphate-activated protein kinase at threonine-172 (AMPKThr172). Hyperglycemia impaired metformin-induced AMPKThr172 activation and enhanced phosphorylation of AMPK at serine-485 (AMPKSer485). Metformin blocked the inhibitory effect of insulin-like growth factor 1 receptor (IGF-1R)/insulin receptor substrate 1 (IRS-1) pathway on TSC-2, and hyperglycemia impaired metformin-induced inhibition of IGF-1R/IRS-1 pathway and modulated the invasiveness of bile duct cancer cells; however, this effect was impaired by hyperglycemia. Conclusions: Metformin has antineoplastic effects in bile duct cancer, and hyperglycemic environment interrupts the effect of metformin. In addition, AMPK and IGF-1R play a key role in the proliferation of bile duct cancer cells.
Collapse
Affiliation(s)
- Jin Lee
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Eun Mi Hong
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Jung Han Kim
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Seoul, Korea
| | - Jang Han Jung
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Dong Hee Koh
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Min Ho Choi
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| | - Sea Hyub Kae
- Division of Gastroenterology, Department of Internal Medicine, Hallym University College of Medicine, Gyeonggi-Do, Korea
| |
Collapse
|
27
|
Yu Q, Zhang M, Qian L, Wen D, Wu G. Luteolin attenuates high glucose-induced podocyte injury via suppressing NLRP3 inflammasome pathway. Life Sci 2019; 225:1-7. [PMID: 30935950 DOI: 10.1016/j.lfs.2019.03.073] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 12/30/2022]
Abstract
AIMS Diabetic nephropathy is a growing health concern, which is reported to be associated with inflammation. Luteolin has been explored for the treatment of some diabetic complications. Although several studies have verified the effect of luteolin on diabetic nephropathy, the mechanism by which the therapeutic effects of luteolin on diabetic nephropathy has not been established. Therefore, we aimed to investigate the effect of luteolin on diabetic nephropathy and its underlying mechanism. MAIN METHODS We used western blot, Real-time PCR, immunofluorescence and flow cytometry to analyze the effects of luteolin on podocyte injury and NOD-like receptor family and pyrin domain-containing protein 3 (NLRP3) inflammasome activation in high glucose (HG) condition. Reactive oxygen species (ROS) generation was measured by flow cytometry and malondialdehyde (MDA) level. To investigate the potential mechanism, we examined cell apoptosis upon transfection of siNLRP3. KEY FINDINGS We showed that luteolin treatment could protect podocyte against HG-induced cell apoptotic and mitochondrial membrane potential collapse. In addition, luteolin significantly reduced NLRP3 inflammasome formation and subsequent interleukin-1β (IL-1β) secretion in HG-induced MPC-5 cells. Interestingly, siNLRP3 abolished the effect of luteolin on cell apoptosis, suggesting that the anti-apoptotic effect was found to be mostly related to NLRP3 inflammasome. SIGNIFICANCE In summary, our data demonstrated the abilities of luteolin to inhibit podocyte injury and NLRP3 inflammasome activation, which could be used in the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Qian Yu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Minda Zhang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Lifen Qian
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Dan Wen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China
| | - Guanzhong Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, PR China.
| |
Collapse
|
28
|
Esmaeili S, Motamedrad M, Hemmati M, Mehrpour O, Khorashadizadeh M. Prevention of kidney cell damage in hyperglycaemia condition by adiponectin. Cell Biochem Funct 2019; 37:148-152. [PMID: 30908696 DOI: 10.1002/cbf.3380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 01/27/2019] [Indexed: 11/07/2022]
Abstract
Adiponectin (APN) is an adipocytokine, secreted from adipose tissue and has anti-inflammatory, anti-ageing, and antidiabetic properties. Hyperglycaemia can damage the renal cells, and mammalian target of rapamycin (mTOR), along with Sirtuin 1 (SIRT1), have an important role in kidney cell response to hyperglycaemia. Therefore, understanding the relationship between adiponectin, mTOR, and SIRT1 proteins is beneficial for deciphering the mechanism of adiponectin function. In this study, Human Embryonic Kidney-293 (HEK-293) cells were cultured under normal and high-glucose condition, with and without APN (1, 10, and 100 ng/mL) for 48 hours. mTOR protein expression was evaluated by western blot analysis, and SIRT1 protein was assessed using ELISA method. To evaluate hyperglycaemia-mediated cytotoxicity, cell viability was determined using MTT assay. Data showed that APN in high dose (100 ng/mL) significantly reduced the expression of mTOR and p-mTOR, increased SIRT1 protein, and also improved cell viability compared with the control high glucose (p ≤ 0.05). According to this results, APN can be useful in preventing renal cell damage, by affecting on the expression of mTOR and SIRT1 proteins, as well as increasing the survival of kidney cells in hyperglycaemia conditions. SIGNIFICANCE OF THE STUDY: Adiponectin triggered mTOR/p-mTOR/SIRT1 pathway and decreased cell death in human kidney cells. Our findings provide preliminary experimental data that support further studies on the potential therapeutic role of adiponectin in diabetes and diabetic-induced metabolic complications.
Collapse
Affiliation(s)
- Sajad Esmaeili
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Maryam Motamedrad
- Department of Biology, Faculty of Science, University of Birjand, Birjand, Iran
| | - Mina Hemmati
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Omid Mehrpour
- Medical Toxicology and Drug Abuse Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Khorashadizadeh
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
29
|
Schulz A, Müller NV, van de Lest NA, Eisenreich A, Schmidbauer M, Barysenka A, Purfürst B, Sporbert A, Lorenzen T, Meyer AM, Herlan L, Witten A, Rühle F, Zhou W, de Heer E, Scharpfenecker M, Panáková D, Stoll M, Kreutz R. Analysis of the genomic architecture of a complex trait locus in hypertensive rat models links Tmem63c to kidney damage. eLife 2019; 8:42068. [PMID: 30900988 PMCID: PMC6478434 DOI: 10.7554/elife.42068] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 03/20/2019] [Indexed: 12/23/2022] Open
Abstract
Unraveling the genetic susceptibility of complex diseases such as chronic kidney disease remains challenging. Here, we used inbred rat models of kidney damage associated with elevated blood pressure for the comprehensive analysis of a major albuminuria susceptibility locus detected in these models. We characterized its genomic architecture by congenic substitution mapping, targeted next-generation sequencing, and compartment-specific RNA sequencing analysis in isolated glomeruli. This led to prioritization of transmembrane protein Tmem63c as a novel potential target. Tmem63c is differentially expressed in glomeruli of allele-specific rat models during onset of albuminuria. Patients with focal segmental glomerulosclerosis exhibited specific TMEM63C loss in podocytes. Functional analysis in zebrafish revealed a role for tmem63c in mediating the glomerular filtration barrier function. Our data demonstrate that integrative analysis of the genomic architecture of a complex trait locus is a powerful tool for identification of new targets such as Tmem63c for further translational investigation. The human kidneys filter the entire volume of the blood about 300 times each day. This ability depends on specialized cells, known as podocytes, which wrap around some of the blood vessels in the kidney. These cells control which molecules leave the blood based on their size. Normally large molecules like proteins are blocked, while smaller molecules including waste products, toxins, excess water and salts pass through into the urine. If this filtration system is damaged, by high blood pressure, for example, it can lead to chronic kidney disease. A hallmark of this disease, often called CKD for short, is high levels of the protein albumin in the urine. Previous studies involving rats with high blood pressure have found several regions of the genome that contribute to high levels of albumin in the urine, including one on chromosome 6. However, this region contains several genes and it was unclear which genes affected the condition. Schulz et al. set out to narrow down the list and find specific genes that might contribute to elevated albumin in the urine of rats with high blood pressure. This search identified the gene for a protein called TMEM63c as a likely candidate. This protein spans the outer membrane of podocyte cells. Analysis of kidney biopsies showed that patients with chronic kidney disease also had low levels of this protein in their podocytes. Further experiments, this time in zebrafish, showed that reducing the activity of the gene for tmem63c led to damaged podocytes and a leakier filter in the kidneys. The results suggest that this gene plays an important role in the integrity of the kidneys filtration barrier. It is possible that faulty versions of this gene are behind some cases of chronic kidney disease. If this proves to be the case, a better understanding of the role of this gene may lead to new treatments for the condition.
Collapse
Affiliation(s)
- Angela Schulz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany
| | - Nicola Victoria Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Electrochemical Signaling in Development and Disease, Berlin, Germany
| | - Nina Anne van de Lest
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Andreas Eisenreich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany
| | - Martina Schmidbauer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany
| | - Andrei Barysenka
- Westfälische Wilhelms University, Genetic Epidemiology, Institute for Human Genetics, Münster, Germany
| | - Bettina Purfürst
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Core Facility Electron Microscopy, Berlin, Germany
| | - Anje Sporbert
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Advanced Light Microscopy, Berlin, Germany
| | - Theodor Lorenzen
- Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany
| | | | - Laura Herlan
- Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany
| | - Anika Witten
- Westfälische Wilhelms University, Genetic Epidemiology, Institute for Human Genetics, Münster, Germany
| | - Frank Rühle
- Westfälische Wilhelms University, Genetic Epidemiology, Institute for Human Genetics, Münster, Germany
| | - Weibin Zhou
- Division of Nephrology, Department of Medicine, Center for Human Disease Modeling, Duke University School of Medicine, Durham, United States
| | - Emile de Heer
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Marion Scharpfenecker
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Daniela Panáková
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Monika Stoll
- Westfälische Wilhelms University, Genetic Epidemiology, Institute for Human Genetics, Münster, Germany.,Department of Biochemistry, Maastricht University, Genetic Epidemiology and Statistical Genetics, Maastricht, The Netherlands
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| |
Collapse
|
30
|
Wang X, Tang D, Zou Y, Wu X, Chen Y, Li H, Chen S, Shi Y, Niu H. A mitochondrial-targeted peptide ameliorated podocyte apoptosis through a HOCl-alb-enhanced and mitochondria-dependent signalling pathway in diabetic rats and in vitro. J Enzyme Inhib Med Chem 2019; 34:394-404. [PMID: 30734599 PMCID: PMC6327984 DOI: 10.1080/14756366.2018.1488697] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mitochondria play important roles in the development of diabetic kidney disease (DKD). The SS peptide is a tetrapeptide that is located and accumulated in the inner mitochondrial membrane; it reduces reactive oxygen species (ROS) and prevents mitochondrial dysfunction. Podocytes are key cellular components in DKD progression. However, whether the SS peptide can exert renal protection through podocytes and the mechanism involved are unknown. In the present study, we explored the mechanisms of the SS peptide on podocyte injury in vivo and in vitro. Compared with the control group, the glomerular podocyte number and expression of WT1 were significantly reduced and TUNEL-positive podocytes were significantly increased in renal tissues in the diabetic rat. These effects were further exacerbated by hypochlorite-modified albumin (HOCl-alb) challenge but prevented by SS-31. In vitro, SS-31 blocked apoptosis in podocyte cell line induced by HOCl-alb. SS-31 prevented oxidative stress and mitochondria-dependent apoptosis signalling by HOCl-alb in vivo and in vitro, as evidenced by the release of cytochrome c (cyt c), binding of apoptosis activated factor-1 (Apaf-1) and caspase-9, and activation of caspases. These data suggest that SS-31 may prevent podocyte apoptosis, exerting renal protection in diabetes mellitus, probably through an apoptosis-related signalling pathway involving oxidative stress and culminating in mitochondria.
Collapse
Affiliation(s)
- Xiaoqiao Wang
- a Division of Nephrology, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Dongdong Tang
- b Division of Nephrology, Huadu District people's Hospital , Southern Medical University , Guangzhou , China
| | - Yaowei Zou
- a Division of Nephrology, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Xiaoyu Wu
- a Division of Nephrology, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Yihua Chen
- a Division of Nephrology, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Hongying Li
- c Special Medical Service Center, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Siqi Chen
- a Division of Nephrology, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Yue Shi
- c Special Medical Service Center, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| | - Hongxin Niu
- c Special Medical Service Center, Zhujiang Hospital , Southern Medical University , Guangzhou , China
| |
Collapse
|
31
|
Szrejder M, Piwkowska A. AMPK signalling: Implications for podocyte biology in diabetic nephropathy. Biol Cell 2019; 111:109-120. [DOI: 10.1111/boc.201800077] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Maria Szrejder
- Mossakowski Medical Research Centre Polish Academy of SciencesLaboratory of Molecular and Cellular Nephrology Gdańsk Poland
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Centre Polish Academy of SciencesLaboratory of Molecular and Cellular Nephrology Gdańsk Poland
| |
Collapse
|
32
|
Polianskyte-Prause Z, Tolvanen TA, Lindfors S, Dumont V, Van M, Wang H, Dash SN, Berg M, Naams JB, Hautala LC, Nisen H, Mirtti T, Groop PH, Wähälä K, Tienari J, Lehtonen S. Metformin increases glucose uptake and acts renoprotectively by reducing SHIP2 activity. FASEB J 2019; 33:2858-2869. [PMID: 30321069 PMCID: PMC6338644 DOI: 10.1096/fj.201800529rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/24/2018] [Indexed: 01/05/2023]
Abstract
Metformin, the first-line drug to treat type 2 diabetes (T2D), inhibits mitochondrial glycerolphosphate dehydrogenase in the liver to suppress gluconeogenesis. However, the direct target and the underlying mechanisms by which metformin increases glucose uptake in peripheral tissues remain uncharacterized. Lipid phosphatase Src homology 2 domain-containing inositol-5-phosphatase 2 (SHIP2) is upregulated in diabetic rodent models and suppresses insulin signaling by reducing Akt activation, leading to insulin resistance and diminished glucose uptake. Here, we demonstrate that metformin directly binds to and reduces the catalytic activity of the recombinant SHIP2 phosphatase domain in vitro. Metformin inhibits SHIP2 in cultured cells and in skeletal muscle and kidney of db/db mice. In SHIP2-overexpressing myotubes, metformin ameliorates reduced glucose uptake by slowing down glucose transporter 4 endocytosis. SHIP2 overexpression reduces Akt activity and enhances podocyte apoptosis, and both are restored to normal levels by metformin. SHIP2 activity is elevated in glomeruli of patients with T2D receiving nonmetformin medication, but not in patients receiving metformin, compared with people without diabetes. Furthermore, podocyte loss in kidneys of metformin-treated T2D patients is reduced compared with patients receiving nonmetformin medication. Our data unravel a novel molecular mechanism by which metformin enhances glucose uptake and acts renoprotectively by reducing SHIP2 activity.-Polianskyte-Prause, Z., Tolvanen, T. A., Lindfors, S., Dumont, V., Van, M., Wang, H., Dash, S. N., Berg, M., Naams, J.-B., Hautala, L. C., Nisen, H., Mirtti, T., Groop, P.-H., Wähälä, K., Tienari, J., Lehtonen, S. Metformin increases glucose uptake and acts renoprotectively by reducing SHIP2 activity.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Kidney Diseases/prevention & control
- Male
- Metformin/pharmacology
- Mice
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
- Podocytes/cytology
- Podocytes/drug effects
- Podocytes/metabolism
- Rats
Collapse
Affiliation(s)
| | | | - Sonja Lindfors
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Vincent Dumont
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mervi Van
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Hong Wang
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Surjya N. Dash
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mika Berg
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | | | - Laura C. Hautala
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Harry Nisen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Central Clinical School, Monash University, Melbourne, Victoria, Australia; and
| | - Kristiina Wähälä
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, Hyvinkää, Finland
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
33
|
Müller T, Kalea AZ, Marquez A, Hsieh I, Haque S, Ye M, Wysocki J, Bader M, Batlle D. Apelinergic system in the kidney: implications for diabetic kidney disease. Physiol Rep 2018; 6:e13939. [PMID: 30548130 PMCID: PMC6288480 DOI: 10.14814/phy2.13939] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/19/2022] Open
Abstract
The bioactive peptides of the apelinergic system and its receptor APJ have been shown to play a protective role in experimental cardiovascular and diabetic kidney disease (DKD). Mechanisms of this renoprotective effect remain to be elucidated. In this study, we examined the localization of APJ within the normal kidney and its kidney expression in the db/db model of DKD. The effect of hyperglycemia and angiotensin II on APJ was examined in cultured podocytes. In the glomerulus, APJ colocalized with podocyte but not endothelial cell markers. In podocytes stimulated with Pyr1 Apelin-13, a change in the phosphorylation status of the signaling proteins, AKT, ERK, and p70S6K, was observed with an increase 15 min after stimulation. Apelin-13 decreased activity of Caspase-3 in podocytes after high glucose treatment reflecting an antiapoptotic effect of APJ stimulation. In podocytes, APJ mRNA was downregulated in high glucose, when compared to normal glucose conditions and exposure to angiotensin II led to a further significant decrease in APJ mRNA. APJ and preproapelin mRNA levels in kidneys from db/db mice were markedly decreased along with decreased tubular APJ protein by western blotting and immunostaining when compared to db/m controls. In conclusion, the apelinergic system is decreased in kidneys from db/db mice. Within the glomerulus, APJ is mainly localized in podocytes and in this cell type its activation by Apelin-13 abolishes the proapoptotic effect of high glucose, suggesting a potential therapeutic role of apelin and emerging agonists with extended half-life for therapy of DKD.
Collapse
Affiliation(s)
- Tilman Müller
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Charité‐Universitätsmedizin BerlinBerlinGermany
| | - Anastasia Z. Kalea
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Institute of Liver and Digestive HealthUniversity College LondonLondonUK
| | - Alonso Marquez
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Ivy Hsieh
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Syed Haque
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Minghao Ye
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Jan Wysocki
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Michael Bader
- Charité‐Universitätsmedizin BerlinBerlinGermany
- Max Delbrück Center for Molecular MedicineBerlinGermany
- German Center for Cardiovascular Research (DZHK), partner site BerlinBerlinGermany
- Berlin Institute of Health (BIH)BerlinGermany
- University of LübeckLübeckGermany
| | - Daniel Batlle
- Department of MedicineDivision of Nephrology and HypertensionFeinberg School of MedicineNorthwestern UniversityChicagoIllinois
| |
Collapse
|
34
|
Wang Q, Sun Y, Li T, Liu L, Zhao Y, Li L, Zhang L, Meng Y. Function of BRD4 in the pathogenesis of high glucose‑induced cardiac hypertrophy. Mol Med Rep 2018; 19:499-507. [PMID: 30483785 PMCID: PMC6297744 DOI: 10.3892/mmr.2018.9681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Diabetic cardiomyopathy is one of the major complications of diabetes, and due to the increasing number of patients with diabetes it is a growing concern. Diabetes-induced cardiomyopathy has a complex pathogenesis and histone deacetylase-mediated epigenetic processes are of prominent importance. The olfactory bromodomain-containing protein 4 (BRD4) is a protein that recognizes and binds acetylated lysine. It has been reported that the high expression of BRD4 is involved in the process of cardiac hypertrophy. The aim of the present study was to investigate the function of BRD4 in the process of high glucose (HG)-induced cardiac hypertrophy, and to clarify whether epigenetic regulation involving BRD4 is an important mechanism. It was revealed that BRD4 expression levels were increased in H9C2 cells following 48 h of HG stimulation. This result was also observed in a diabetic rat model. Furthermore, HG stimulation resulted in the upregulation of the myocardial hypertrophy marker, atrial natriuretic peptide, the cytoskeletal protein α-actin and fibrosis-associated genes including transforming growth factor-β, SMAD family member 3, connective tissue growth factor and collagen, type 1, α1. However, administration of the specific BRD4 inhibitor JQ1 (250 nM) for 48 h reversed this phenomenon. Furthermore, protein kinase B (AKT) phosphorylation was activated by HG stimulation and suppressed by JQ1. In conclusion, BRD4 serves an important role in the pathogenesis of HG-induced cardiomyocyte hypertrophy through the AKT pathway.
Collapse
Affiliation(s)
- Qian Wang
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuxin Sun
- Department of Otorhinolaryngology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tianshu Li
- Department of Functional Science Experiment Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lianqin Liu
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yunxia Zhao
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liyuan Li
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ling Zhang
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Meng
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
35
|
Scheen AJ. Effects of glucose-lowering agents on surrogate endpoints and hard clinical renal outcomes in patients with type 2 diabetes. DIABETES & METABOLISM 2018; 45:110-121. [PMID: 30477733 DOI: 10.1016/j.diabet.2018.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022]
Abstract
Diabetic kidney disease (DKD) represents an enormous burden in patients with type 2 diabetes mellitus (T2DM). Preclinical studies using most glucose-lowering agents have suggested renal-protective effects, but the proposed mechanisms of renoprotection have yet to be defined, and the promising results from experimental studies remain to be translated into human clinical findings to improve the prognosis of patients at risk of DKD. Also, it is important to distinguish effects on surrogate endpoints, such as decreases in albuminuria and estimated glomerular filtration rate (eGFR), and hard clinical endpoints, such as progression to end-stage renal disease (ESRD) and death from renal causes. Data regarding insulin therapy are surprisingly scarce, and it is nearly impossible to separate the effects of better glucose control from those of insulin per se, whereas favourable preclinical data with metformin, thiazolidinediones and dipeptidyl peptidase (DPP)-4 inhibitors are plentiful, and positive effects have been observed in clinical studies, at least for surrogate endpoints. The most favourable renal results have been reported with glucagon-like peptide-1 receptor agonists (GLP-1RAs) and sodium-glucose cotransporter type-2 inhibitors (SGLT2is). Significant reductions in both albuminuria and eGFR decline have been reported with these classes of glucose-lowering medications compared with placebo and other glucose-lowering agents. Moreover, in large prospective cardiovascular outcome trials using composite renal outcomes as secondary endpoints, both GLP-1RAs and SGLT2is added to standard care reduced renal outcomes combining persistent macro-albuminuria, doubling of serum creatinine, progression to ESRD and kidney-related death; however, to date, only SGLT2is have been clearly shown to reduce such hard clinical outcomes. Yet, as the renoprotective effects of SGLT2is and GLP-1RAs appear to be independent of glucose-lowering activity, the underlying mechanisms are still a matter of debate. For this reason, further studies with renal outcomes as primary endpoints are now awaited in T2DM patients at high risk of DKD, including trials evaluating the potential add-on benefits of combined GLP-1RA-SGLT2i therapies.
Collapse
Affiliation(s)
- A J Scheen
- Division of Clinical Pharmacology, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU de Liège, Liège, Belgium.
| |
Collapse
|
36
|
Huang YS, Wang SH, Chen SM, Lee JA. Metabolic profiling of metformin treatment for low-level Pb-induced nephrotoxicity in rat urine. Sci Rep 2018; 8:14587. [PMID: 30275489 PMCID: PMC6167321 DOI: 10.1038/s41598-018-32501-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/06/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease is a worldwide problem, and Pb contamination is a potential risk factor. Since current biomarkers are not sensitive for the diagnosis of Pb-induced nephrotoxicity, novel biomarkers are needed. Metformin has both hypoglycaemic effects and reno-protection ability. However, its mechanism of action is unknown. We aimed to discover the early biomarkers for the diagnosis of low-level Pb-induced nephrotoxicity and understand the mechanism of reno-protection of metformin. Male Wistar rats were randomly divided into control, Pb, Pb + ML, Pb + MH and MH groups. Pb (250 ppm) was given daily via drinking water. Metformin (50 or 100 mg/kg/d) was orally administered. Urine was analysed by nuclear magnetic resonance (NMR)-based metabolomics coupled with multivariate statistical analysis, and potential biomarkers were subsequently quantified. The results showed that Pb-induced nephrotoxicity was closely correlated with the elevation of 5-aminolevulinic acid, D-lactate and guanidinoacetic acid in urine. After co-treatment with metformin, 5-aminolevulinic acid and D-lactate were decreased. This is the first demonstration that urinary 5-aminolevulinic acid, D-lactate and guanidinoacetic acid could be early biomarkers of low-level Pb-induced nephrotoxicity in rats. The reno-protection of metformin might be attributable to the reduction of D-lactate excretion.
Collapse
Affiliation(s)
- Yu-Shen Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing St., Taipei, Taiwan
| | - Shwu-Huey Wang
- Core Facility Center, Department of Research Development, Taipei Medical University, 250 Wuxing St., Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Taipei Medical University, 250 Wuxing St., Taipei, Taiwan
| | - Shih-Ming Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing St., Taipei, Taiwan.
| | - Jen-Ai Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing St., Taipei, Taiwan.
| |
Collapse
|
37
|
De Broe M, Kajbaf F, Lalau JD. Renoprotective Effects of Metformin. Nephron Clin Pract 2017; 138:261-274. [DOI: 10.1159/000481951] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/03/2017] [Indexed: 12/18/2022] Open
|
38
|
Lu H, Li Y, Zhang T, Liu M, Chi Y, Liu S, Shi Y. Salidroside Reduces High-Glucose-Induced Podocyte Apoptosis and Oxidative Stress via Upregulating Heme Oxygenase-1 (HO-1) Expression. Med Sci Monit 2017; 23:4067-4076. [PMID: 28831032 PMCID: PMC5580518 DOI: 10.12659/msm.902806] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Hyperglycemia is one of the most dangerous factors causing diabetic nephropathy. Salidroside is considered to have the effects of reducing oxidative stress damage and improving cell viability. This study was performed to investigate whether and how salidroside reduces high-glucose (HG)-induced apoptosis in mouse podocytes. Material/Methods We examined whether salidroside could decrease HG-induced podocyte oxidative stress and podocyte apoptosis in vitro. The potential signaling pathways were also investigated. Podocytes (immortalized mouse epithelial cells) were treated with normal glucose (5.5 mM) as control or HG (30 mM), and then exposed to salidroside treatment. Results HG enhanced the generation of intracellular reactive oxygen species (ROS) and apoptosis in podocytes. Salidroside reduced HG-induced apoptosis-related consequences via promoting HO-1 expression. Salidroside increased the expression level of phosphorylated Akt (p-Akt) and phosphorylated ILK (p-ILK), p-JNK, and p-ERK and localization of Nrf-2. JNK inhibitor and ILK inhibitor decreased HO-1 expression to different degrees. Moreover, specific siRNAs of ILK, Nrf-2, and HO-1, and inhibitors of HO-1 and ILK significantly increased ROS generation and Caspase9/3 expression in the presence of salidroside and HG. Conclusions The results suggest that salidroside reduces HG-induced ROS generation and apoptosis and improves podocytes viability by upregulating HO-1 expression. ILK/Akt, JNK, ERK1/2, p38 MAPK, and Nrf-2 are involved in salidroside-decreased podocyte apoptosis in HG condition.
Collapse
Affiliation(s)
- Hua Lu
- Department of Nephrology, The 3rd Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland)
| | - Ying Li
- Department of Nephrology, The 3rd Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland)
| | - Tao Zhang
- Department of Nephrology, The 3rd Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland)
| | - Maodong Liu
- Department of Nephrology, The 3rd Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland)
| | - Yanqing Chi
- Department of Nephrology, The 3rd Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland)
| | - Shuxia Liu
- Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland).,Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Yonghong Shi
- Key Laboratory of Kidney Diseases of Hebei Province, Shijiazhuang, Hebei, China (mainland).,Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
39
|
Chen X, Qin Y, Zhou T, Jiang L, Lei F, Qin H, Zhang L, Zhou Z. The potential role of retinoic acid receptor α on glomerulosclerosis in rats and podocytes injury is associated with the induction of MMP2 and MMP9. Acta Biochim Biophys Sin (Shanghai) 2017; 49:669-679. [PMID: 28645189 DOI: 10.1093/abbs/gmx066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Indexed: 02/05/2023] Open
Abstract
Retinoic acid receptor α (RARα) plays a crucial role in kidney disease. However, the underlying mechanisms in glomerulosclerosis (GS) is still not clear. The roles of RARα in an adriamycin (ADR)-induced GS rat model and in ADR-induced podocyte injury in vitro were investigated. RARα was over-expressed in GS rats, and serum, urine and kidney samples were collected to detect the induction of the expression of the receptor. RARα expression was inhibited and/or over-expressed in cultured podocytes following injury, as demonstrated by morphometric assays, cell toxicity, and matrix metalloproteinase (MMP) enzymatic activity. RARα displayed a renoprotective role in GS rats, resulting in a lower GS index, podocyte foot process fusion, and proteinuria, reduced serum creatinine and blood urea nitrogen. Further experiments indicated that RARα inhibited the accumulation of TGF-β1, α-smooth muscle actin, collagen IV, and fibronectin, while it induced MMP2 and MMP9 excessive expression in podocytes in vitro. RARα improved the renal function and attenuated the progression of GS that was associated with the over-expression of MMP2 and MMP9.
Collapse
Affiliation(s)
- Xiuping Chen
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yuanhan Qin
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Ling Jiang
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Fengying Lei
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - He Qin
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Lei Zhang
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhiqiang Zhou
- Department of Pediatrics Nephrology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
40
|
Leppert U, Gillespie A, Orphal M, Böhme K, Plum C, Nagorsen K, Berkholz J, Kreutz R, Eisenreich A. The impact of α-Lipoic acid on cell viability and expression of nephrin and ZNF580 in normal human podocytes. Eur J Pharmacol 2017; 810:1-8. [PMID: 28606850 DOI: 10.1016/j.ejphar.2017.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 02/04/2023]
Abstract
Human podocytes (hPC) are essential for maintaining normal kidney function and dysfunction or loss of hPC play a pivotal role in the manifestation and progression of chronic kidney diseases including diabetic nephropathy. Previously, α-Lipoic acid (α-LA), a licensed drug for treatment of diabetic neuropathy, was shown to exhibit protective effects on diabetic nephropathy in vivo. However, the effect of α-LA on hPC under non-diabetic conditions is unknown. Therefore, we analyzed the impact of α-LA on cell viability and expression of nephrin and zinc finger protein 580 (ZNF580) in normal hPC in vitro. Protein analyses were done via Western blot techniques. Cell viability was determined using a functional assay. hPC viability was dynamically modulated via α-LA stimulation in a concentration-dependent manner. This was associated with reduced nephrin and ZNF580 expression and increased nephrin phosphorylation in normal hPC. Moreover, α-LA reduced nephrin and ZNF580 protein expression via 'kappa-light-chain-enhancer' of activated B-cells (NF-κB) inhibition. These data demonstrate that low α-LA had no negative influence on hPC viability, whereas, high α-LA concentrations induced cytotoxic effects on normal hPC and reduced nephrin and ZNF580 expression via NF-κB inhibition. These data provide first novel information about potential cytotoxic effects of α-LA on hPC under non-diabetic conditions.
Collapse
Affiliation(s)
- Ulrike Leppert
- Charité-Universitätsmedizin Berlin, CC02, Institut für Physiologie, Berlin, Germany
| | - Allan Gillespie
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Miriam Orphal
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Karen Böhme
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Claudia Plum
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Kaj Nagorsen
- Charité-Universitätsmedizin Berlin, CC02, Institut für Physiologie, Berlin, Germany
| | - Janine Berkholz
- Charité-Universitätsmedizin Berlin, CC02, Institut für Physiologie, Berlin, Germany
| | - Reinhold Kreutz
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany
| | - Andreas Eisenreich
- Charité-Universitätsmedizin Berlin, CC04, Institut für Klinische Pharmakologie und Toxikologie, Berlin, Germany.
| |
Collapse
|
41
|
Wang Y, Zhang J, Zhang L, Gao P, Wu X. Adiponectin attenuates high glucose-induced apoptosis through the AMPK/p38 MAPK signaling pathway in NRK-52E cells. PLoS One 2017; 12:e0178215. [PMID: 28542560 PMCID: PMC5444659 DOI: 10.1371/journal.pone.0178215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022] Open
Abstract
Excessive apoptosis of proximal tubule cell is closely related to the development of diabetes. Recent evidence suggests that adiponectin (ADPN) protects cells from high glucose induced apoptosis. However, the precise mechanisms remain poorly understood. We sought to investigate the role of p38 mitogen-activated protein kinase (p38 MAPK) and AMP activated protein kinase (AMPK) in anti-apoptotic of adiponectin under high glucose condition in rat tubular NRK-52E cells. Cells were cultured in constant and oscillating high glucose media with or without recombinant rat adiponectin for 48 h. Cell counting kit-8 (CCK-8) was used to detect cell viability, flow cytometry and Hoechst Staining were applied to investigate cell apoptosis, and western blotting was used to examine protein expression, such as phospho-AMPK and phospho-p38MAPK. Exposure to oscillating high glucose exerted lower cell viability and higher early apoptosis than constant high glucose, which were both partially prevented by adiponectin. Further studies revealed that adiponectin suppressed p38MAPK phosphorylation, but led to an increase in AMPK α phosphorylation. Compared to stable high glucose group, blockage of p38MAPK cascade with SB203580 attenuated apoptosis significantly, but failed to affect the phosphorylation level of AMPK. While AMPK inhibitor, Compound C, increased apoptosis and remarkably inhibited the p38MAPK phosphorylation. Adiponectin exert a crucial protective role against apoptosis induced by high glucose via AMPK/p38MAPK pathway.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juan Zhang
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lian Zhang
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ping Gao
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoyan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- * E-mail:
| |
Collapse
|
42
|
Wang S, Wang J, Zhang Z, Miao H. Decreased miR-128 and increased miR-21 synergistically cause podocyte injury in sepsis. J Nephrol 2017; 30:543-550. [PMID: 28497421 DOI: 10.1007/s40620-017-0405-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/27/2017] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Glomerular podocytes are injured in sepsis. We studied, in a sepsis patient, whether microRNAs (miRNAs) play a role in the podocyte injury. METHODS Podocytes were cultured and treated with lipopolysaccharide (LPS). Filtration barrier function of podocyte was analyzed with albumin influx assay. Nephrin level was analyzed with reverse transcription polymerase chain reaction (RT-PCR) and western blot. MiRNAs were detected using miRNAs PCR Array and in situ hybridization. MiRNA target sites were evaluated with luciferase reporter assays. RESULTS LPS impaired the filtration barrier function of podocytes. MiR-128 level was decreased and miR-21 level was increased in podocytes in vitro and in the sepsis patient. The decrease in miR-128 was sufficient to induce the loss of nephrin and the impairment of filtration barrier function, while the increase of miR-21 exacerbated the process. Snail and phosphatase and tensin homolog (PTEN) were identified as the targets of miR-128 and miR-21. Decreased miR-128 induced Snail expression, and the increased miR-21 stabilized Snail by regulating the PTEN/Akt/GSK3β pathway. Supplementation of miR-128 and inhibition of miR-21 suppressed Snail expression and prevented the podocyte injury induced by LPS. CONCLUSION Our study suggests that decreased miR-128 and increased miR-21 synergistically cause podocyte injury and are the potential therapeutic targets in sepsis.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Emergency, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Jun Wang
- Department of Emergency, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zengdi Zhang
- Department of Emergency, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Hongjun Miao
- Department of Emergency, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
43
|
Du L, Wang M, Kang Y, Li B, Guo M, Cheng Z, Bi C. Prognostic role of metformin intake in diabetic patients with colorectal cancer: An updated qualitative evidence of cohort studies. Oncotarget 2017; 8:26448-26459. [PMID: 28103573 PMCID: PMC5432271 DOI: 10.18632/oncotarget.14688] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/29/2016] [Indexed: 12/23/2022] Open
Abstract
Several observational studies have shown that metformin can modify the risk and survival of colorectal cancer (CRC) in patients with diabetes mellitus, although the magnitude of this relationship has not been determined. We conducted an updated systematic review and meta-analysis to analyze the association between metformin and CRC mortality and searched relevant databases up to July 2016. The primary outcome was overall survival (OS). Secondary outcomes were cancer-specific survival (CS) and disease-free survival (DFS). Summary hazard ratios (HRs) were calculated using a random-effects model. Seventeen studies enrolling 269,417 participants were eligible for inclusion. Comparing with non-metformin users in diabetic CRC patients, the summary HRs for OS in metformin users were 0.69 (95% CI, 0.61-0.77). Subgroup analyses stratified by the study characteristics and sensitivity analysis by the trim-and-fill method (adjusted HR 0.77, 95% CI, 0.67-0.87) confirmed the robustness of the results. However, significant OS benefit was noted in patients with stage II and III disease. Five studies reported the CRC prognosis for CS and three for DFS; metformin intake was significantly associated with patient CS (HR 0.75, 95% CI, 0.59-0.94), but not DFS (HR 0.38, 95% CI, 0.13-1.17). Our findings suggest that metformin intake is associated with improved survival outcomes in terms of OS and CS in CRC patients with diabetes, particular for OS in stage II and stage III patients. Further studies should be conducted to determine CRC survival between metformin use and patient specific clinical and molecular profiles.
Collapse
Affiliation(s)
- Lili Du
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingli Wang
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingying Kang
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Li
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Min Guo
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhifeng Cheng
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Changlong Bi
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
44
|
Metadherin facilitates podocyte apoptosis in diabetic nephropathy. Cell Death Dis 2016; 7:e2477. [PMID: 27882943 PMCID: PMC5260885 DOI: 10.1038/cddis.2016.335] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/23/2016] [Accepted: 09/23/2016] [Indexed: 02/01/2023]
Abstract
Apoptosis, one of the major causes of podocyte loss, has been reported to have a vital role in diabetic nephropathy (DN) pathogenesis, and understanding the mechanisms underlying the regulation of podocyte apoptosis is crucial. Metadherin (MTDH) is an important oncogene, which is overexpressed in most cancers and responsible for apoptosis, metastasis, and poor patient survival. Here we show that the expression levels of Mtdh and phosphorylated p38 mitogen-activated protein kinase (MAPK) are significantly increased, whereas those of the microRNA-30 family members (miR-30s) are considerably reduced in the glomeruli of DN rat model and in high glucose (HG)-induced conditionally immortalized mouse podocytes (MPC5). These levels are positively correlated with podocyte apoptosis rate. The inhibition of Mtdh expression, using small interfering RNA, but not Mtdh overexpression, was shown to inhibit HG-induced MPC5 apoptosis and p38 MAPK pathway, and Bax and cleaved caspase 3 expression. This was shown to be similar to the effects of p38 MAPK inhibitor (SB203580). Furthermore, luciferase assay results demonstrated that Mtdh represents the target of miR-30s. Transient transfection experiments, using miR-30 microRNA (miRNA) inhibitors, led to the increase in Mtdh expression and induced the apoptosis of MPC5, whereas the treatment with miR-30 miRNA mimics led to the reduction in Mtdh expression and apoptosis of HG-induced MPC5 cells in comparison with their respective controls. Our results demonstrate that Mtdh is a potent modulator of podocyte apoptosis, and that it represents the target of miR-30 miRNAs, facilitating podocyte apoptosis through the activation of HG-induced p38 MAPK-dependent pathway.
Collapse
|
45
|
Jin Y, Liu S, Ma Q, Xiao D, Chen L. Berberine enhances the AMPK activation and autophagy and mitigates high glucose-induced apoptosis of mouse podocytes. Eur J Pharmacol 2016; 794:106-114. [PMID: 27887947 DOI: 10.1016/j.ejphar.2016.11.037] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022]
Abstract
High glucose concentration can induce injury of podocytes and berberine has a potent activity against diabetic nephropathy. However, whether and how berberine can inhibit high glucose-mediated injury of podocytes have not been clarified. This study tested the effect of berberine on high glucose-mediated apoptosis and the AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR) activation and autophagy in podocytes. The results indicated that berberine significantly mitigated high glucose-decreased cell viability, and nephrin and podocin expression as well as apoptosis in mouse podocytes. Berberine significantly increased the AMPK activation and mitigated high glucose and/or the AMPK inhibitor, compound C-mediated mTOR activation and apoptosis in podocytes. Berberine significantly enhanced the AMPK activation and protected from high glucose-induced apoptosis in the AMPK-silencing podocytes. Furthermore, berberine significantly increased the high glucose-elevated Unc-51-like autophagy-activating kinase 1 (ULK1) S317/S555 phosphorylation, Beclin-1 expression, the ratios of LC3II to LC3I expression and the numbers of autophagosomes, but reduced ULK1 S757 phosphorylation in podocytes. In addition, berberine significantly attenuated compound C-mediated inhibition of autophagy in podocytes. The protective effect of berberine on high glucose-induced podocyte apoptosis was significantly mitigated by pre-treatment with 3-methyladenine or bafilomycin A1. Collectively, berberine enhanced autophagy and protected from high glucose-induced injury in podocytes by promoting the AMPK activation. Our findings may provide new insights into the molecular mechanisms underlying the anti-diabetic nephropathy effect of berberine and may aid in design of new therapies for intervention of diabetic nephropathy.
Collapse
Affiliation(s)
- Yingli Jin
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Xinmin Street 126, Changchun 130021, China
| | - Shuping Liu
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Xinmin Street 126, Changchun 130021, China
| | - Qingshan Ma
- Department of Pediatrics, the First Bethune Hospital of Jilin University, Jilin University, Xinmin Street 71, Changchun 130021, China
| | - Dong Xiao
- Academy of Translational Medicine, the First Bethune Hospital of Jilin University, Jilin University, Xinmin Street 71, Changchun 130021, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Xinmin Street 126, Changchun 130021, China.
| |
Collapse
|
46
|
Lin C, Zhang M, Zhang Y, Yang K, Hu J, Si R, Zhang G, Gao B, Li X, Xu C, Li C, Hao Q, Guo W. Helix B surface peptide attenuates diabetic cardiomyopathy via AMPK-dependent autophagy. Biochem Biophys Res Commun 2016; 482:665-671. [PMID: 27865838 DOI: 10.1016/j.bbrc.2016.11.091] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Erythropoietin (EPO) has been reported to exert protective effects on a host of damaged tissues. However, the erythropoietic effect of this hormone can result in high risks of thrombosis, stroke, and hypertension, remarkably limiting the clinical use of EPO. Helix B surface peptide (HBSP) is a small peptide derived from the helix-B domain of EPO. Surprisingly, HBSP retains the tissue protective properties of EPO without altering the hematocrit. Thus, we evaluated the possible role of HBSP on diabetic cardiomyopathy. METHODS Diabetes was induced in mice by intraperitoneal injections of streptozocin (STZ). Mice were randomly treated with normal saline or HBSP. Cardiac function, fibrosis, apoptosis, and myocardial mitochondrial morphology were examined. For in vitro experiments, H9C2 myoblast cells were randomly grouped as normal glucose (NG, 5 mM), NG+HBSP (100 nM), high glucose (HG, 33 mM), HG+HBSP (100 nM), HG+HBSP+3-methyladenine (3-MA, 10 mM), HG+rapamycin (Rapa, 100 nM), and HG+HBSP+Compound C (CC, 10 mM). Autophagosomes, LC3 dots, apoptosis and mitochondria membrane potential (MMP) of H9C2 cells were examined.The expressions of LC3, p62, p-AMPK (Thr172) and p-mTOR (Ser2448) were examined by Western blot. RESULTS HBSP markedly improved cardiac function, attenuated cardiac interstitial fibrosis, inhibited myocardial apoptosis, and ameliorated mitochondrial ultrastructure in mice with diabetic cardiomyopathy. HG reduced autophagy in H9C2 cells. HBSP enhanced autophagy in HG-treated H9C2 cells. HBSP reduced the apoptosis index of HG-treated H9C2 cells. HBSP increased the MMP of HG-treated H9C2 cells. HBSP increased the levels of p-AMPK (Thr172), and reduced p-mTOR (Ser2448) in HG-treated H9C2 cells, and the increase of p-AMPK (Thr172) was accompanied by the stimulation of autophagy. Autophagy inhibitor 3-MA and AMPK inhibitor CC mitigated HBSP-induced beneficial effect, whereas autophagy inducer Rapa alleviated the HG-induced cell apoptosis. CONCLUSIONS HBSP attenuates diabetic cardiomyopathy via autophagy mediated by AMPK-dependent pathway. HBSP may be a potential therapeutic intervention for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Chen Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mingming Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kejian Yang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianqiang Hu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Rui Si
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guoyong Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Beilei Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chennian Xu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qimeng Hao
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenyi Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
47
|
Wang H, Wang J, Qu H, Wei H, Ji B, Yang Z, Wu J, He Q, Luo Y, Liu D, Duan Y, Liu F, Deng H. In vitro and in vivo inhibition of mTOR by 1,25-dihydroxyvitamin D 3 to improve early diabetic nephropathy via the DDIT4/TSC2/mTOR pathway. Endocrine 2016; 54:348-359. [PMID: 27395420 DOI: 10.1007/s12020-016-0999-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023]
Abstract
We investigated whether 1,25-dihydroxy-vitamin D3 (1,25(OH)2D3) could improve early diabetic nephropathy through the DNA-damage-inducible transcript 4/tuberous sclerosis 2/mammalian target of rapamycin pathway. Rat mesangial cells were cultured in media containing normal glucose or high glucose and were treated with or without 1,25(OH)2D3. Mesangial cells proliferation was measured. Streptozotocin-induced diabetic rats were injected intravenously with a recombinant lentivirus against the rat vitamin D receptor gene. Urinary and serum albumin, fasting plasma glucose, serum triglyceride, total cholesterol, calcium, parathyroid hormone and serum 25-dihydroxy-vitamin D (25(OH)D) levels, mean glomerular volume, glomerular basement membrane thickness and total kidney volume were determined. The expressions of vitamin D receptor, DNA-damage-inducible transcript 4, and mammalian target of rapamycin were measured. 1,25(OH)2D3 inhibited the proliferation of mesangial cells induced by hyperglycemia. 1,25(OH)2D3 also significantly reduced albumin excretion, mean glomerular volume, glomerular basement membrane, and total kidney volume in rats with diabetic nephropathy. The expression of DNA-damage-inducible transcript 4 was elevated by 1,25(OH)2D3 treatment. The phosphorylation of mammalian target of rapamycin was reduced by 1,25(OH)2D3 treatment. Vitamin D receptor gene silencing blocked all of the above results. The current study demonstrates that 1,25(OH)2D3 can effectively inhibit mesangial cells proliferation induced by hyperglycemia, thus suppressing the development of diabetic nephropathy. This study also shows that the nephron-protective effect of 1,25(OH)2D3 occurs partly through the DDIT4/TSC2/mTOR pathway.
Collapse
Affiliation(s)
- Hang Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jianmin Wang
- Department of Nephrology, Chongqing Armed Corps Police Hospital, Chongqing, P.R. China
| | - Hua Qu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Huili Wei
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Baolan Ji
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Zesong Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jing Wu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Qin He
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Yuanyuan Luo
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Dan Liu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Yang Duan
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Fang Liu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Huacong Deng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China.
| |
Collapse
|