1
|
Wang XL, Li L, Meng X. Interplay between the Redox System and Renal Tubular Transport. Antioxidants (Basel) 2024; 13:1156. [PMID: 39456410 PMCID: PMC11505102 DOI: 10.3390/antiox13101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The kidney plays a critical role in maintaining the homeostasis of body fluid by filtration of metabolic wastes and reabsorption of nutrients. Due to the overload, a vast of energy is required through aerobic metabolism, which inevitably leads to the generation of reactive oxygen species (ROS) in the kidney. Under unstressed conditions, ROS are counteracted by antioxidant systems and maintained at low levels, which are involved in signal transduction and physiological processes. Accumulating evidence indicates that the reduction-oxidation (redox) system interacts with renal tubular transport. Redox imbalance or dysfunction of tubular transport leads to renal disease. Here, we discuss the ROS and antioxidant systems in the kidney and outline the metabolic dysfunction that is a common feature of renal disease. Importantly, we describe the key molecules involved in renal tubular transport and their relationship to the redox system and, finally, summarize the impact of their dysregulation on the pathogenesis and progression of acute and chronic kidney disease.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Lianjian Li
- Department of Vascular Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Hubei Academy of Chinese Medicine, Wuhan 430061, China;
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
2
|
D’Haese S, Claes L, Jaeken E, Deluyker D, Evens L, Heeren E, Haesen S, Vastmans L, Lambrichts I, Wouters K, Schalkwijk CG, Hansen D, Eijnde BO, Bito V. Pyridoxamine Alleviates Cardiac Fibrosis and Oxidative Stress in Western Diet-Induced Prediabetic Rats. Int J Mol Sci 2024; 25:8508. [PMID: 39126079 PMCID: PMC11312841 DOI: 10.3390/ijms25158508] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Individuals with type 2 diabetes mellitus (T2DM) are at an increased risk for heart failure, yet preventive cardiac care is suboptimal in this population. Pyridoxamine (PM), a vitamin B6 analog, has been shown to exert protective effects in metabolic and cardiovascular diseases. In this study, we aimed to investigate whether PM limits adverse cardiac remodeling and dysfunction in rats who develop T2DM. Male rats received a standard chow diet or Western diet (WD) for 18 weeks to induce prediabetes. One WD group received additional PM (1 g/L) via drinking water. Glucose tolerance was assessed with a 1 h oral glucose tolerance test. Cardiac function was evaluated using echocardiography and hemodynamic measurements. Histology on left ventricular (LV) tissue was performed. Treatment with PM prevented the increase in fasting plasma glucose levels compared to WD-fed rats (p < 0.05). LV cardiac dilation tended to be prevented using PM supplementation. In LV tissue, PM limited an increase in interstitial collagen deposition (p < 0.05) seen in WD-fed rats. PM tended to decrease 3-nitrotyrosine and significantly lowered 4-hydroxynonenal content compared to WD-fed rats. We conclude that PM alleviates interstitial fibrosis and oxidative stress in the hearts of WD-induced prediabetic rats.
Collapse
Affiliation(s)
- Sarah D’Haese
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Lisa Claes
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Eva Jaeken
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Dorien Deluyker
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Lize Evens
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Ellen Heeren
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Sibren Haesen
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Lotte Vastmans
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Ivo Lambrichts
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Kristiaan Wouters
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Casper G. Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Dominique Hansen
- UHasselt, Faculty of Rehabilitation Sciences, REVAL Rehabilitation Research Centre, Agoralaan, 3590 Diepenbeek, Belgium;
- Department of Cardiology, Heart Centre Hasselt, Jessa Hospital, Stadsomvaart 11, 3500 Hasselt, Belgium
| | - BO Eijnde
- SMRc-Sports Medicine Research Center, BIOMED-Biomedical Research Institute, Faculty of Medicine & Life Sciences, Hasselt University, 3590 Diepenbeek, Belgium;
- Division of Sport Science, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Virginie Bito
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| |
Collapse
|
3
|
Wang M, Wang S, Tang HP, Li JY, Zhang ZJ, Yang BY, Kuang HX. Buddleja officinalis Maxim.: A review of its botany, ethnopharmacology, phytochemistry, pharmacology, and therapeutic potential for ophthalmic diseases. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116993. [PMID: 37541402 DOI: 10.1016/j.jep.2023.116993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Buddleja officinalis Maxim. (B. officinalis), commonly known as "Menghua" "Yangerduo" is a widely recognized traditional herbal medicine in China, Korea, and Vietnam. For thousands of years, it has been used to treat dry eye disease, conjunctivitis, keratitis, eye ulcers, eye pain, cough, asthma, hemoptysis, and other medical conditions. AIM OF THE REVIEW This review article aims to provide a concise summary of the botany, ethnopharmacology, phytochemistry, pharmacology, medicinal potential, and application of B. officinalis in treating ophthalmic diseases and critically evaluates the existing literature to establish a scientific basis for its reasonable utilization and further investigation. MATERIALS AND METHODS The information reviewed in this study was collected from various electronic resources, including the Web of Science, PubMed, and Google Scholar. RESULTS To date, 80 structurally diverse compounds have been isolated and characterized from B. officinalis, primarily flavonoids, phenylethanoids, triterpenoids, and monoterpenes. Extracts and compounds derived from B. officinalis have been reported to possess broad pharmacological effects including anti-dry eye disease, anti-inflammation, anti-oxidation, anti-diabetes, anti-obesity, improving osteoporosis and treatment of skin diseases. This review provides a reference for the future studies on of B. officinalis. CONCLUSIONS As a natural medicinal plant, B. officinalis is worthy of further development in botany, ethnopharmacology, phytochemistry, pharmacology, and therapeutic potential for ophthalmic diseases. Although some components have demonstrated multiple pharmacological activities, their mechanisms of action remain unclear. Further studies on the underlying molecular basis and mechanism of action are warranted.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Shuang Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Hai-Peng Tang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Jia-Yan Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Zhao-Jiong Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Bing-You Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Hai-Xue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China.
| |
Collapse
|
4
|
Butcko AJ, Putman AK, Mottillo EP. The Intersection of Genetic Factors, Aberrant Nutrient Metabolism and Oxidative Stress in the Progression of Cardiometabolic Disease. Antioxidants (Basel) 2024; 13:87. [PMID: 38247511 PMCID: PMC10812494 DOI: 10.3390/antiox13010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/06/2023] [Accepted: 01/07/2024] [Indexed: 01/23/2024] Open
Abstract
Cardiometabolic disease (CMD), which encompasses metabolic-associated fatty liver disease (MAFLD), chronic kidney disease (CKD) and cardiovascular disease (CVD), has been increasing considerably in the past 50 years. CMD is a complex disease that can be influenced by genetics and environmental factors such as diet. With the increased reliance on processed foods containing saturated fats, fructose and cholesterol, a mechanistic understanding of how these molecules cause metabolic disease is required. A major pathway by which excessive nutrients contribute to CMD is through oxidative stress. In this review, we discuss how oxidative stress can drive CMD and the role of aberrant nutrient metabolism and genetic risk factors and how they potentially interact to promote progression of MAFLD, CVD and CKD. This review will focus on genetic mutations that are known to alter nutrient metabolism. We discuss the major genetic risk factors for MAFLD, which include Patatin-like phospholipase domain-containing protein 3 (PNPLA3), Membrane Bound O-Acyltransferase Domain Containing 7 (MBOAT7) and Transmembrane 6 Superfamily Member 2 (TM6SF2). In addition, mutations that prevent nutrient uptake cause hypercholesterolemia that contributes to CVD. We also discuss the mechanisms by which MAFLD, CKD and CVD are mutually associated with one another. In addition, some of the genetic risk factors which are associated with MAFLD and CVD are also associated with CKD, while some genetic risk factors seem to dissociate one disease from the other. Through a better understanding of the causative effect of genetic mutations in CMD and how aberrant nutrient metabolism intersects with our genetics, novel therapies and precision approaches can be developed for treating CMD.
Collapse
Affiliation(s)
- Andrew J. Butcko
- Hypertension and Vascular Research Division, Henry Ford Hospital, 6135 Woodward Avenue, Detroit, MI 48202, USA; (A.J.B.); (A.K.P.)
- Department of Physiology, Wayne State University, 540 E. Canfield Street, Detroit, MI 48202, USA
| | - Ashley K. Putman
- Hypertension and Vascular Research Division, Henry Ford Hospital, 6135 Woodward Avenue, Detroit, MI 48202, USA; (A.J.B.); (A.K.P.)
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 784 Wilson Road, East Lansing, MI 48823, USA
| | - Emilio P. Mottillo
- Hypertension and Vascular Research Division, Henry Ford Hospital, 6135 Woodward Avenue, Detroit, MI 48202, USA; (A.J.B.); (A.K.P.)
- Department of Physiology, Wayne State University, 540 E. Canfield Street, Detroit, MI 48202, USA
| |
Collapse
|
5
|
Gui Y, Palanza Z, Fu H, Zhou D. Acute kidney injury in diabetes mellitus: Epidemiology, diagnostic, and therapeutic concepts. FASEB J 2023; 37:e22884. [PMID: 36943403 PMCID: PMC10602403 DOI: 10.1096/fj.202201340rr] [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: 08/15/2022] [Revised: 02/16/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023]
Abstract
Acute kidney injury (AKI) and diabetes mellitus (DM) are public health problems that cause a high socioeconomic burden worldwide. In recent years, the landscape of AKI etiology has shifted: Emerging evidence has demonstrated that DM is an independent risk factor for the onset of AKI, while an alternative perspective considers AKI as a bona fide complication of DM. Therefore, it is necessary to systematically characterize the features of AKI in DM. In this review, we summarized the epidemiology of AKI in DM. While focusing on circulation- and tissue-specific microenvironment changes after DM, we described the active cellular and molecular mechanisms of increased kidney susceptibility to AKI under DM stress. We also reviewed the current diagnostic and therapeutic strategies for AKI in DM recommended in the clinic. Updated recognition of the epidemiology, pathophysiology, diagnosis, and medications of AKI in DM is believed to reveal a path to mitigate the frequency of AKI and DM comorbidity that will ultimately improve the quality of life in DM patients.
Collapse
Affiliation(s)
- Yuan Gui
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Zachary Palanza
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Haiyan Fu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| |
Collapse
|
6
|
Zhang C, Li H, Wang S. Common gene signatures and molecular mechanisms of diabetic nephropathy and metabolic syndrome. Front Public Health 2023; 11:1150122. [PMID: 37143982 PMCID: PMC10151256 DOI: 10.3389/fpubh.2023.1150122] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023] Open
Abstract
Background Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. Multiple metabolic toxicities, redox stress, and endothelial dysfunction contribute to the development of diabetic glomerulosclerosis and DN. Metabolic syndrome (MetS) is a pathological state in which the body's ability to process carbohydrates, fats, and proteins is compromised because of metabolic disorders, resulting in redox stress and renal remodeling. However, a causal relationship between MetS and DN has not been proven. This study aimed to provide valuable information for the clinical diagnosis and treatment of MetS with DN. Methods Here, transcriptome data of DN and MetS patients were obtained from the Gene Expression Omnibus database, and seven potential biomarkers were screened using bioinformatics analysis. In addition, the relationship between these marker genes and metabolism and immune infiltration was explored. Among the identified marker genes, the relationship between PLEKHA1 and the cellular process, oxidative phosphorylation (OXPHOS), in DN was further investigated through single-cell analysis. Results We found that PLEKHA1 may represent an important biomarker that perhaps initiates DN by activating B cells, proximal tubular cells, distal tubular cells, macrophages, and endothelial cells, thereby inducing OXPHOS in renal monocytes. Conclusion Overall, our findings can aid in further investigation of the effects of drug treatment on single cells of patients with diabetes to validate PLEKHA1 as a therapeutic target and to inform the development of targeted therapies.
Collapse
|
7
|
Ferreira A, Rivera A, Wohlgemuth JG, Dlott JS, Snyder LM, Alper SL, Romero JR. Dysregulated Erythroid Mg2+ Efflux in Type 2 Diabetes. Front Cell Dev Biol 2022; 10:861644. [PMID: 35445032 PMCID: PMC9013827 DOI: 10.3389/fcell.2022.861644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/09/2022] [Indexed: 12/23/2022] Open
Abstract
Hyperglycemia is associated with decreased Mg2+ content in red blood cells (RBC), but mechanisms remain unclear. We characterized the regulation of Mg2+ efflux by glucose in ex vivo human RBC. We observed that hemoglobin A1C (HbA1C) values correlated with Na+-dependent Mg2+ efflux (Na+/Mg2+ exchange) and inversely correlated with cellular Mg content. Treatment of cells with 50 mM D-glucose, but not with sorbitol, lowered total cellular Mg (2.2 ± 0.1 to 2.0 ± 0.1 mM, p < 0.01) and enhanced Na+/Mg2+ exchange activity [0.60 ± 0.09 to 1.12 ± 0.09 mmol/1013 cell × h (flux units, FU), p < 0.05]. In contrast, incubation with selective Src family kinase inhibitors PP2 or SU6656 reduced glucose-stimulated exchange activation (p < 0.01). Na+/Mg2+ exchange activity was also higher in RBC from individuals with type 2 diabetes (T2D, 1.19 ± 0.13 FU) than from non-diabetic individuals (0.58 ± 0.05 FU, p < 0.01). Increased Na+/Mg2+ exchange activity in RBC from T2D subjects was associated with lower intracellular Mg content. Similarly increased exchange activity was evident in RBC from the diabetic db/db mouse model as compared to its non-diabetic control (p < 0.03). Extracellular exposure of intact RBC from T2D subjects to recombinant peptidyl-N-glycosidase F (PNGase F) reduced Na+/Mg2+ exchange activity from 0.98 ± 0.14 to 0.59 ± 0.13 FU (p < 0.05) and increased baseline intracellular Mg content (1.8 ± 0.1 mM) to normal values (2.1 ± 0.1 mM, p < 0.05). These data suggest that the reduced RBC Mg content of T2D RBC reflects enhanced RBC Na+/Mg2+ exchange subject to regulation by Src family kinases and by the N-glycosylation state of one or more membrane proteins. The data extend our understanding of dysregulated RBC Mg2+ homeostasis in T2D.
Collapse
Affiliation(s)
- Ana Ferreira
- Interdisciplinary Centre of Social Sciences (CICS.NOVA), Faculty of Social Sciences and Humanities (NOVA FCSH), Lisbon, Portugal
| | - Alicia Rivera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- *Correspondence: Alicia Rivera,
| | | | | | | | - Seth L. Alper
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Jose R. Romero
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Vallon V, Nakagawa T. Renal Tubular Handling of Glucose and Fructose in Health and Disease. Compr Physiol 2021; 12:2995-3044. [PMID: 34964123 PMCID: PMC9832976 DOI: 10.1002/cphy.c210030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The proximal tubule of the kidney is programmed to reabsorb all filtered glucose and fructose. Glucose is taken up by apical sodium-glucose cotransporters SGLT2 and SGLT1 whereas SGLT5 and potentially SGLT4 and GLUT5 have been implicated in apical fructose uptake. The glucose taken up by the proximal tubule is typically not metabolized but leaves via the basolateral facilitative glucose transporter GLUT2 and is returned to the systemic circulation or used as an energy source by distal tubular segments after basolateral uptake via GLUT1. The proximal tubule generates new glucose in metabolic acidosis and the postabsorptive phase, and fructose serves as an important substrate. In fact, under physiological conditions and intake, fructose taken up by proximal tubules is primarily utilized for gluconeogenesis. In the diabetic kidney, glucose is retained and gluconeogenesis enhanced, the latter in part driven by fructose. This is maladaptive as it sustains hyperglycemia. Moreover, renal glucose retention is coupled to sodium retention through SGLT2 and SGLT1, which induces secondary deleterious effects. SGLT2 inhibitors are new anti-hyperglycemic drugs that can protect the kidneys and heart from failing independent of kidney function and diabetes. Dietary excess of fructose also induces tubular injury. This can be magnified by kidney formation of fructose under pathological conditions. Fructose metabolism is linked to urate formation, which partially accounts for fructose-induced tubular injury, inflammation, and hemodynamic alterations. Fructose metabolism favors glycolysis over mitochondrial respiration as urate suppresses aconitase in the tricarboxylic acid cycle, and has been linked to potentially detrimental aerobic glycolysis (Warburg effect). © 2022 American Physiological Society. Compr Physiol 12:2995-3044, 2022.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, California, USA,Department of Pharmacology, University of California San Diego, La Jolla, California, USA,VA San Diego Healthcare System, San Diego, California, USA,Correspondence to and
| | - Takahiko Nakagawa
- Division of Nephrology, Rakuwakai-Otowa Hospital, Kyoto, Japan,Correspondence to and
| |
Collapse
|
9
|
Ahangarpour A, Oroojan AA, Khorsandi L, Kouchak M, Badavi M. Hyperglycemia-induced oxidative stress in isolated proximal tubules of mouse: the in vitro effects of myricitrin and its solid lipid nanoparticle. Arch Physiol Biochem 2021; 127:422-428. [PMID: 31368364 DOI: 10.1080/13813455.2019.1647250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/18/2022]
Abstract
CONTEXT The hyperglycemia (Hyper) induces oxidative stress in kidney tubular cells. Myricitrin (Myr) has an antioxidant effect along with low bioavailability. OBJECTIVE The present research investigated the effects of Myr and its solid lipid nanoparticles (SLN) on isolated proximal tubules exposed to the hyperglycemic condition. MATERIALS AND METHODS In this experimental study, the proximal tubules of mice were dissected by the microdissection method and the tubules were prepared for experimental or Real Time-PCR measurement. RESULTS The malondialdehyde level, transforming growth factor-β, nuclear factor kappa B and Bax genes expression increased in Hyper and decreased in Hyper + Myr and its SLN-treated groups compared to Hyper. Superoxide dismutase, total antioxidant capacity, the viability of proximal tubules and Bcl-2 gene expression decreased in untreated Hyper and increased in all treatment groups compared to Hyper. CONCLUSION The hyperglycemia-induced oxidative disorder, inflammation and apoptosis in proximal tubules and that administrating Myr and its SLN improved them.
Collapse
Affiliation(s)
- Akram Ahangarpour
- Department of Physiology, Faculty of Medicine, Diabetes Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Akbar Oroojan
- Department of Physiology, Faculty of Medicine, Cellular and Molecular Research Center, Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Layasadat Khorsandi
- Department of Anatomical Sciences, Faculty of Medicine, Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Department of Pharmaceutics, Faculty of Pharmacy, Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
10
|
Astragaloside IV attenuates high glucose-induced EMT by inhibiting the TGF-β/Smad pathway in renal proximal tubular epithelial cells. Biosci Rep 2021; 40:225214. [PMID: 32515466 PMCID: PMC7313447 DOI: 10.1042/bsr20190987] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022] Open
Abstract
In the present study, we examined the molecular mechanism of astragaloside IV (AS-IV) in high glucose (HG)-induced epithelial-to-mesenchymal transition (EMT) in renal proximal tubular epithelial cells (PTCs). NRK-52E cell viability and apoptosis were determined by the cell counting kit-8 (CCK-8) assay and flow cytometric analysis, respectively. Expressions of E-cadherin, N-cadherin, vimentin, and occludin were measured by Western blot, and those of E-cadherin and N-cadherin were additionally measured by immunofluorescence analysis. Transforming growth factor-β1 (TGF-β1) and α-smooth muscle actin (α-SMA) expressions were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. The expressions of Smad2, Smad3, phosphorylated-Smad2 (p-Smad2), and p-Smad3 were measured using Western blot. We found that AS-IV could recover NRK-52E cell viability and inhibit HG-induced cell apoptosis. TGF-β1, α-SMA, Smad2, Smad3, p-Smad2, and p-Smad3 expressions were decreased in the AS-IV-treated groups compared with the HG group. Moreover, the expressions of E-cadherin and occludin were remarkably up-regulated and those of N-cadherin and vimentin were down-regulated in the AS-IV-treated groups compared with the HG group. Interestingly, the TGF-β1 activator SRI-011381 hydrochloride had an antagonistic effect to AS-IV on HG-induced EMT behavior. In conclusion, AS-IV attenuates HG-induced EMT by inhibiting the TGF-β/Smad pathway in renal PTCs.
Collapse
|
11
|
Piani F, Reinicke T, Borghi C, Tommerdahl KL, Cara-Fuentes G, Johnson RJ, Bjornstad P. Acute Kidney Injury in Pediatric Diabetic Kidney Disease. Front Pediatr 2021; 9:668033. [PMID: 34211943 PMCID: PMC8239177 DOI: 10.3389/fped.2021.668033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Diabetic kidney disease (DKD) is a common complication of type 1 and 2 diabetes and often presents during adolescence and young adulthood. Given the growing incidence of both type 1 and type 2 diabetes in children and adolescents, DKD represents a significant public health problem. Acute kidney injury (AKI) in youth with diabetes is strongly associated with risk of DKD development. This review will summarize the epidemiology and pathophysiology of AKI in children with diabetes, the relationship between AKI and DKD, and the potential therapeutic interventions. Finally, we will appraise the impact of the recent COVID-19 infection pandemic on AKI in children with diabetes.
Collapse
Affiliation(s)
- Federica Piani
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Section of Pediatric Endocrinology, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Medicine and Surgery Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Trenton Reinicke
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Claudio Borghi
- Department of Medicine and Surgery Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Kalie L Tommerdahl
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Section of Pediatric Endocrinology, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Gabriel Cara-Fuentes
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Section of Pediatric Nephrology, Department of Pediatrics, Children's Hospital Colorado, Aurora, CO, United States
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Petter Bjornstad
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Section of Pediatric Endocrinology, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
12
|
Vallon V. Glucose transporters in the kidney in health and disease. Pflugers Arch 2020; 472:1345-1370. [PMID: 32144488 PMCID: PMC7483786 DOI: 10.1007/s00424-020-02361-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The kidneys filter large amounts of glucose. To prevent the loss of this valuable fuel, the tubular system of the kidney, particularly the proximal tubule, has been programmed to reabsorb all filtered glucose. The machinery involves the sodium-glucose cotransporters SGLT2 and SGLT1 on the apical membrane and the facilitative glucose transporter GLUT2 on the basolateral membrane. The proximal tubule also generates new glucose, particularly in the post-absorptive phase but also to enhance bicarbonate formation and maintain acid-base balance. The glucose reabsorbed or formed by the proximal tubule is primarily taken up into peritubular capillaries and returned to the systemic circulation or provided as an energy source to further distal tubular segments that take up glucose by basolateral GLUT1. Recent studies provided insights on the coordination of renal glucose reabsorption, formation, and usage. Moreover, a better understanding of renal glucose transport in disease states is emerging. This includes the kidney in diabetes mellitus, when renal glucose retention becomes maladaptive and contributes to hyperglycemia. Furthermore, enhanced glucose reabsorption is coupled to sodium retention through the sodium-glucose cotransporter SGLT2, which induces secondary deleterious effects. As a consequence, SGLT2 inhibitors are new anti-hyperglycemic drugs that can protect the kidneys and heart from failing. Recent studies discovered unique roles for SGLT1 with implications in acute kidney injury and glucose sensing at the macula densa. This review discusses established and emerging concepts of renal glucose transport, and outlines the need for a better understanding of renal glucose handling in health and disease.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
13
|
Abstract
Significance: Fibrosis is a stereotypic, multicellular tissue response to diverse types of injuries that fundamentally result from a failure of cell/tissue regeneration. This complex tissue remodeling response disrupts cellular/matrix composition and homeostatic cell-cell interactions, leading to loss of normal tissue architecture and progressive loss of organ structure/function. Fibrosis is a common feature of chronic diseases that may affect the lung, kidney, liver, and heart. Recent Advances: There is emerging evidence to support a combination of genetic, environmental, and age-related risk factors contributing to susceptibility and/or progression of fibrosis in different organ systems. A core pathway in fibrogenesis involving these organs is the induction and activation of nicotinamide adenine dinucleotide phosphate oxidase (NOX) family enzymes. Critical Issues: We explore current pharmaceutical approaches to targeting NOX enzymes, including repurposing of currently U.S. Food and Drug Administration (FDA)-approved drugs. Specific inhibitors of various NOX homologs will aid establishing roles of NOXs in the various organ fibroses and potential efficacy to impede/halt disease progression. Future Directions: The discovery of novel and highly specific NOX inhibitors will provide opportunities to develop NOX inhibitors for treatment of fibrotic pathologies.
Collapse
Affiliation(s)
- Karen Bernard
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
14
|
Xu N, Jiang S, Persson PB, Persson EAG, Lai EY, Patzak A. Reactive oxygen species in renal vascular function. Acta Physiol (Oxf) 2020; 229:e13477. [PMID: 32311827 DOI: 10.1111/apha.13477] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are produced by the aerobic metabolism. The imbalance between production of ROS and antioxidant defence in any cell compartment is associated with cell damage and may play an important role in the pathogenesis of renal disease. NADPH oxidase (NOX) family is the major ROS source in the vasculature and modulates renal perfusion. Upregulation of Ang II and adenosine activates NOX via AT1R and A1R in renal microvessels, leading to superoxide production. Oxidative stress in the kidney prompts renal vascular remodelling and increases preglomerular resistance. These are key elements in hypertension, acute and chronic kidney injury, as well as diabetic nephropathy. Renal afferent arterioles (Af), the primary resistance vessel in the kidney, fine tune renal hemodynamics and impact on blood pressure. Vice versa, ROS increase hypertension and diabetes, resulting in upregulation of Af vasoconstriction, enhancement of myogenic responses and change of tubuloglomerular feedback (TGF), which further promotes hypertension and diabetic nephropathy. In the following, we highlight oxidative stress in the function and dysfunction of renal hemodynamics. The renal microcirculatory alterations brought about by ROS importantly contribute to the pathophysiology of kidney injury, hypertension and diabetes.
Collapse
Affiliation(s)
- Nan Xu
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Pontus B. Persson
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | | | - En Yin Lai
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Andreas Patzak
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
15
|
Vallon V, Thomson SC. The tubular hypothesis of nephron filtration and diabetic kidney disease. Nat Rev Nephrol 2020; 16:317-336. [PMID: 32152499 DOI: 10.1038/s41581-020-0256-y] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 02/08/2023]
Abstract
Kidney size and glomerular filtration rate (GFR) often increase with the onset of diabetes, and elevated GFR is a risk factor for the development of diabetic kidney disease. Hyperfiltration mainly occurs in response to signals passed from the tubule to the glomerulus: high levels of glucose in the glomerular filtrate drive increased reabsorption of glucose and sodium by the sodium-glucose cotransporters SGLT2 and SGLT1 in the proximal tubule. Passive reabsorption of chloride and water also increases. The overall capacity for proximal reabsorption is augmented by growth of the proximal tubule, which (alongside sodium-glucose cotransport) further limits urinary glucose loss. Hyperreabsorption of sodium and chloride induces tubuloglomerular feedback from the macula densa to increase GFR. In addition, sodium-glucose cotransport by SGLT1 on macula densa cells triggers the production of nitric oxide, which also contributes to glomerular hyperfiltration. Although hyperfiltration restores sodium and chloride excretion it imposes added physical stress on the filtration barrier and increases the oxygen demand to drive reabsorption. Tubular growth is associated with the development of a senescence-like molecular signature that sets the stage for inflammation and fibrosis. SGLT2 inhibitors attenuate the proximal reabsorption of sodium and glucose, normalize tubuloglomerular feedback signals and mitigate hyperfiltration. This tubule-centred model of diabetic kidney physiology predicts the salutary effect of SGLT2 inhibitors on hard renal outcomes, as shown in large-scale clinical trials.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA. .,Department of Pharmacology, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Healthcare System, San Diego, CA, USA.
| | - Scott C Thomson
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA.,VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
16
|
García-Pastor C, Benito-Martínez S, Moreno-Manzano V, Fernández-Martínez AB, Lucio-Cazaña FJ. Mechanism and Consequences of The Impaired Hif-1α Response to Hypoxia in Human Proximal Tubular HK-2 Cells Exposed to High Glucose. Sci Rep 2019; 9:15868. [PMID: 31676796 PMCID: PMC6825166 DOI: 10.1038/s41598-019-52310-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 10/16/2019] [Indexed: 01/01/2023] Open
Abstract
Renal hypoxia and loss of proximal tubular cells (PTC) are relevant in diabetic nephropathy. Hypoxia inhibits hypoxia-inducible factor-1α (HIF-1α) degradation, which leads to cellular adaptive responses through HIF-1-dependent activation of gene hypoxia-responsive elements (HRE). However, the diabetic microenvironment represses the HIF-1/HRE response in PTC. Here we studied the mechanism and consequences of impaired HIF-1α regulation in human proximal tubular HK-2 cells incubated in hyperglycemia. Inhibition at different levels of the canonical pathway of HIF-1α degradation did not activate the HIF-1/HRE response under hyperglycemia, except when proteasome was inhibited. Further studies suggested that hyperglycemia disrupts the interaction of HIF-1α with Hsp90, a known cause of proteasomal degradation of HIF-1α. Impaired HIF-1α regulation in cells exposed to hyperglycemic, hypoxic diabetic-like milieu led to diminished production of vascular endothelial growth factor-A and inhibition of cell migration (responses respectively involved in tubular protection and repair). These effects, as well as impaired HIF-1α regulation, were reproduced in normoglycemia in HK-2 cells incubated with microparticles released by HK-2 cells exposed to diabetic-like milieu. In summary, these results highlight the role of proteasome-dependent mechanisms of HIF-1α degradation on diabetes-induced HK-2 cells dysfunction and suggest that cell-derived microparticles may mediate negative effects of the diabetic milieu on PTC.
Collapse
Affiliation(s)
- Coral García-Pastor
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain.
| | - Selma Benito-Martínez
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | | |
Collapse
|
17
|
Jacob KA, Leaf DE. Prevention of Cardiac Surgery-Associated Acute Kidney Injury: A Review of Current Strategies. Anesthesiol Clin 2019; 37:729-749. [PMID: 31677688 PMCID: PMC7644277 DOI: 10.1016/j.anclin.2019.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Acute kidney injury is a common and often severe postoperative complication after cardiac surgery, and is associated with poor short-term and long-term outcomes. Numerous randomized controlled trials have been conducted to investigate various strategies for prevention of cardiac surgery-associated acute kidney injury. Unfortunately, most trials that have been conducted to date have been negative. However, encouraging results have been demonstrated with preoperative administration of corticosteroids, leukocyte filtration, and administration of inhaled nitric oxide intraoperatively, and implementation of a Kidney Disease: Improving Global Outcomes bundle of care approach postoperatively. These findings require validation in large, multicenter trials.
Collapse
Affiliation(s)
- Kirolos A Jacob
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Mail Stop E03.511, PO Box 85500, Utrecht 3508 GA, the Netherlands.
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, 75 Francis Street, Medial Research Building Room MR416B, Boston, MA 02115, USA
| |
Collapse
|
18
|
Bashir SO. Concomitant administration of resveratrol and insulin protects against diabetes mellitus type-1-induced renal damage and impaired function via an antioxidant-mediated mechanism and up-regulation of Na +/K +-ATPase. Arch Physiol Biochem 2019; 125:104-113. [PMID: 29436859 DOI: 10.1080/13813455.2018.1437752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study investigated if a combination of resveratrol (RES) and insulin could reverse type 1 diabetic mellitus-induced (T1DM) nephropathy and illustrates mechanism of action. Rats were divided into six groups (n = 10/group) as follows: control, control + RES (20 mg/kg), T1DM, T1DM + RES, T1DM + insulin (1 U/g), and T1DM + RES + insulin and treated for eight weeks. While individual administrations of both drugs significantly but partially restored renal function and cortex architectures, combination therapy of both RES and insulin produced the maximum improvements. Mechanism of actions revealed a synergist effect of both drugs due to hypoglycaemic effect of insulin and the ability of both drugs to increase renal cortex antioxidant enzymes activities, inhibit lipid peroxidation, and up-regulate Na+/K+-ATPase, independent of each others. In conclusion, these data suggest the combined therapy with insulin and RES could provide an excellent combined drug therapy against T1DM-induced nephropathy.
Collapse
Affiliation(s)
- Salah O Bashir
- a Department of Physiology, College of Medicine , King Khalid University , Abha , Saudi Arabia
| |
Collapse
|
19
|
Sunilkumar S, Ford SM. Elevated glucose concentration in culture media decreases membrane trafficking of SGLT2 in LLC-PK 1 cells via a cAMP/PKA-dependent pathway. Am J Physiol Cell Physiol 2019; 316:C913-C924. [PMID: 30943059 DOI: 10.1152/ajpcell.00433.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Na+-dependent glucose reabsorption in the renal proximal tubule is dynamically regulated by changes in blood glucose levels. There is, however, a disparity in reports studying the relationship between hyperglycemia and Na+-glucose-linked transporter (SGLT) function and expression. Similarly, manipulation of the glucose content in growth media of cultured renal cells has been shown to influence SGLT activity. In this investigation, SGLT activity was significantly lower in proximal tubule LLC-PK1 cells cultured in medium containing 17.5 than 5 mM glucose. α-Methyl d-glucopyranoside (AMG) transport kinetics showed reduced apparent Vmax and Km in cells grown in 17.5 mM glucose. SGLT2 was identified as the isoform responsible for glucose transport, and protein expression analyses showed decreased apical membrane localization of SGLT2 in cells grown in 17.5 mM glucose, explaining the reduced activity. Multiple signaling pathways have been implicated in regulation of SGLT activity and trafficking. Elevated media glucose decreased intracellular cAMP and PKA activation, leading to decreased SGLT2 trafficking into the plasma membrane, which was reversed after treatment with 1 µM forskolin. The effects of media glucose on SGLT activity were found to be dependent on p38 MAPK activation due to PKA-mediated signaling. Glucose-modulated AMG uptake is reversible and was associated with altered SGLT2 membrane trafficking and cAMP alterations. In summary, elevated glucose concentrations in culture medium decrease SGLT activity in LLC-PK1 cells by reducing membrane trafficking of SGLT2 via decreasing intracellular cAMP, resulting in a lowered PKA-dependent phosphorylation of p38 MAPK.
Collapse
Affiliation(s)
| | - Sue M Ford
- Department of Pharmaceutical Sciences, St. John's University , Queens, New York
| |
Collapse
|
20
|
Pavlakou P, Dounousi E, Roumeliotis S, Eleftheriadis T, Liakopoulos V. Oxidative Stress and the Kidney in the Space Environment. Int J Mol Sci 2018; 19:ijms19103176. [PMID: 30326648 PMCID: PMC6214023 DOI: 10.3390/ijms19103176] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022] Open
Abstract
In space, the special conditions of hypogravity and exposure to cosmic radiation have substantial differences compared to terrestrial circumstances, and a multidimensional impact on the human body and human organ functions. Cosmic radiation provokes cellular and gene damage, and the generation of reactive oxygen species (ROS), leading to a dysregulation in the oxidants–antioxidants balance, and to the inflammatory response. Other practical factors contributing to these dysregulations in space environment include increased bone resorption, impaired anabolic response, and even difficulties in detecting oxidative stress in blood and urine samples. Enhanced oxidative stress affects mitochondrial and endothelial functions, contributes to reduced natriuresis and the development of hypertension, and may play an additive role in the formation of kidney stones. Finally, the composition of urine protein excretion is significantly altered, depicting possible tubular dysfunction.
Collapse
Affiliation(s)
- Paraskevi Pavlakou
- Department of Nephrology, Medical School, University of Ioannina, 45110 Ioannina, Greece.
| | - Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, 45110 Ioannina, Greece.
| | - Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece.
| | - Theodoros Eleftheriadis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece.
| | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece.
| |
Collapse
|
21
|
Taub M. Gene Level Regulation of Na,K-ATPase in the Renal Proximal Tubule Is Controlled by Two Independent but Interacting Regulatory Mechanisms Involving Salt Inducible Kinase 1 and CREB-Regulated Transcriptional Coactivators. Int J Mol Sci 2018; 19:E2086. [PMID: 30021947 PMCID: PMC6073390 DOI: 10.3390/ijms19072086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022] Open
Abstract
For many years, studies concerning the regulation of Na,K-ATPase were restricted to acute regulatory mechanisms, which affected the phosphorylation of Na,K-ATPase, and thus its retention on the plasma membrane. However, in recent years, this focus has changed. Na,K-ATPase has been established as a signal transducer, which becomes part of a signaling complex as a consequence of ouabain binding. Na,K-ATPase within this signaling complex is localized in caveolae, where Na,K-ATPase has also been observed to regulate Inositol 1,4,5-Trisphosphate Receptor (IP3R)-mediated calcium release. This latter association has been implicated as playing a role in signaling by G Protein Coupled Receptors (GPCRs). Here, the consequences of signaling by renal effectors that act via such GPCRs are reviewed, including their regulatory effects on Na,K-ATPase gene expression in the renal proximal tubule (RPT). Two major types of gene regulation entail signaling by Salt Inducible Kinase 1 (SIK1). On one hand, SIK1 acts so as to block signaling via cAMP Response Element (CRE) Binding Protein (CREB) Regulated Transcriptional Coactivators (CRTCs) and on the other hand, SIK1 acts so as to stimulate signaling via the Myocyte Enhancer Factor 2 (MEF2)/nuclear factor of activated T cell (NFAT) regulated genes. Ultimate consequences of these pathways include regulatory effects which alter the rate of transcription of the Na,K-ATPase β1 subunit gene atp1b1 by CREB, as well as by MEF2/NFAT.
Collapse
Affiliation(s)
- Mary Taub
- Biochemistry Dept., Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main Street, Suite 4902, Buffalo, NY 14203, USA.
| |
Collapse
|
22
|
Yu SMW, Bonventre JV. Acute Kidney Injury and Progression of Diabetic Kidney Disease. Adv Chronic Kidney Dis 2018; 25:166-180. [PMID: 29580581 DOI: 10.1053/j.ackd.2017.12.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 12/23/2022]
Abstract
Diabetic kidney disease, commonly termed diabetic nephropathy (DN), is the most common cause of end-stage kidney disease (ESKD) worldwide. The characteristic histopathology of DN includes glomerular basement membrane thickening, mesangial expansion, nodular glomerular sclerosis, and tubulointerstitial fibrosis. Diabetes is associated with a number of metabolic derangements, such as reactive oxygen species overproduction, hypoxic state, mitochondrial dysfunction, and inflammation. In the past few decades, our knowledge of DN has advanced considerably although much needs to be learned. The traditional paradigm of glomerulus-centered pathophysiology has expanded to the tubule-interstitium, the immune response and inflammation. Biomarkers of proximal tubule injury have been shown to correlate with DN progression, independent of traditional glomerular injury biomarkers such as albuminuria. In this review, we summarize mechanisms of increased susceptibility to acute kidney injury in diabetes mellitus and the roles played by many kidney cell types to facilitate maladaptive responses leading to chronic and end-stage kidney disease.
Collapse
|
23
|
Yao M, Gao F, Wang X, Shi Y, Liu S, Duan H. Nox4 is involved in high glucose-induced apoptosis in renal tubular epithelial cells via Notch pathway. Mol Med Rep 2017; 15:4319-4325. [DOI: 10.3892/mmr.2017.6516] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 02/22/2017] [Indexed: 11/06/2022] Open
|
24
|
Gonzalez-Vicente A, Garvin JL. Effects of Reactive Oxygen Species on Tubular Transport along the Nephron. Antioxidants (Basel) 2017; 6:antiox6020023. [PMID: 28333068 PMCID: PMC5488003 DOI: 10.3390/antiox6020023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/15/2017] [Accepted: 03/18/2017] [Indexed: 12/17/2022] Open
Abstract
Reactive oxygen species (ROS) are oxygen-containing molecules naturally occurring in both inorganic and biological chemical systems. Due to their high reactivity and potentially damaging effects to biomolecules, cells express a battery of enzymes to rapidly metabolize them to innocuous intermediaries. Initially, ROS were considered by biologists as dangerous byproducts of respiration capable of causing oxidative stress, a condition in which overproduction of ROS leads to a reduction in protective molecules and enzymes and consequent damage to lipids, proteins, and DNA. In fact, ROS are used by immune systems to kill virus and bacteria, causing inflammation and local tissue damage. Today, we know that the functions of ROS are not so limited, and that they also act as signaling molecules mediating processes as diverse as gene expression, mechanosensation, and epithelial transport. In the kidney, ROS such as nitric oxide (NO), superoxide (O₂-), and their derivative molecules hydrogen peroxide (H₂O₂) and peroxynitrite (ONO₂-) regulate solute and water reabsorption, which is vital to maintain electrolyte homeostasis and extracellular fluid volume. This article reviews the effects of NO, O₂-, ONO₂-, and H₂O₂ on water and electrolyte reabsorption in proximal tubules, thick ascending limbs, and collecting ducts, and the effects of NO and O₂- in the macula densa on tubuloglomerular feedback.
Collapse
Affiliation(s)
- Agustin Gonzalez-Vicente
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1113AAD, Argentina.
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
25
|
Liu J, Yan Y, Nie Y, Shapiro JI. Na/K-ATPase Signaling and Salt Sensitivity: The Role of Oxidative Stress. Antioxidants (Basel) 2017; 6:E18. [PMID: 28257114 PMCID: PMC5384181 DOI: 10.3390/antiox6010018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Other than genetic regulation of salt sensitivity of blood pressure, many factors have been shown to regulate renal sodium handling which contributes to long-term blood pressure regulation and have been extensively reviewed. Here we present our progress on the Na/K-ATPase signaling mediated sodium reabsorption in renal proximal tubules, from cardiotonic steroids-mediated to reactive oxygen species (ROS)-mediated Na/K-ATPase signaling that contributes to experimental salt sensitivity.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Yanling Yan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
26
|
The Na+-D-glucose cotransporters SGLT1 and SGLT2 are targets for the treatment of diabetes and cancer. Pharmacol Ther 2017; 170:148-165. [DOI: 10.1016/j.pharmthera.2016.10.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Yan Y, Shapiro AP, Mopidevi BR, Chaudhry MA, Maxwell K, Haller ST, Drummond CA, Kennedy DJ, Tian J, Malhotra D, Xie ZJ, Shapiro JI, Liu J. Protein Carbonylation of an Amino Acid Residue of the Na/K-ATPase α1 Subunit Determines Na/K-ATPase Signaling and Sodium Transport in Renal Proximal Tubular Cells. J Am Heart Assoc 2016; 5:e003675. [PMID: 27613772 PMCID: PMC5079028 DOI: 10.1161/jaha.116.003675] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/12/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND We have demonstrated that cardiotonic steroids, such as ouabain, signaling through the Na/K-ATPase, regulate sodium reabsorption in the renal proximal tubule. By direct carbonylation modification of the Pro222 residue in the actuator (A) domain of pig Na/K-ATPase α1 subunit, reactive oxygen species are required for ouabain-stimulated Na/K-ATPase/c-Src signaling and subsequent regulation of active transepithelial (22)Na(+) transport. In the present study we sought to determine the functional role of Pro222 carbonylation in Na/K-ATPase signaling and sodium handling. METHODS AND RESULTS Stable pig α1 knockdown LLC-PK1-originated PY-17 cells were rescued by expressing wild-type rat α1 and rat α1 with a single mutation of Pro224 (corresponding to pig Pro222) to Ala. This mutation does not affect ouabain-induced inhibition of Na/K-ATPase activity, but abolishes the effects of ouabain on Na/K-ATPase/c-Src signaling, protein carbonylation, Na/K-ATPase endocytosis, and active transepithelial (22)Na(+) transport. CONCLUSIONS Direct carbonylation modification of Pro224 in the rat α1 subunit determines ouabain-mediated Na/K-ATPase signal transduction and subsequent regulation of renal proximal tubule sodium transport.
Collapse
Affiliation(s)
- Yanling Yan
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV
| | - Anna P Shapiro
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Brahma R Mopidevi
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Muhammad A Chaudhry
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV
| | - Kyle Maxwell
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | | | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Jiang Tian
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Zi-Jian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Joseph I Shapiro
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Jiang Liu
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
28
|
Wahby EA, Abo Elnasr MM, Eissa MI, Mahmoud SM. Perioperative glycemic control in diabetic patients undergoing coronary artery bypass graft surgery. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jescts.2016.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
29
|
Ku SK, Bae JS. Baicalin, baicalein and wogonin inhibits high glucose-induced vascular inflammation in vitro and in vivo. BMB Rep 2016; 48:519-24. [PMID: 25739393 PMCID: PMC4641236 DOI: 10.5483/bmbrep.2015.48.9.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Indexed: 12/29/2022] Open
Abstract
Vascular inflammatory process has been suggested to play a key role in initiation and progression of atherosclerosis, a major complication of diabetes mellitus. Thus, in this study, we attempted to determine whether three structurally related polyphenols found in the Chinese herb Huang Qui, namely baicalin, baicalein, and wogonin, can suppress vascular inflammatory processes induced by high glucose (HG) in human umbilical vein endothelial cells (HUVECs) and mice. Data showed that HG induced markedly increased vascular permeability, monocyte adhesion, expressions of cell adhesion molecules (CAMs), formation of reactive oxygen species (ROS) and activation of nuclear factor (NF)-κB. Remarkably, all of the above mentioned vascular inflammatory effects of HG were attenuated by pretreatment with baicalin, baicalein, and wogonin. Vascular inflammatory responses induced by HG are critical events underlying development of various diabetic complications, therefore, our results suggest that baicalin, baicalein, and wogonin may have significant therapeutic benefits against diabetic complications and atherosclerosis.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
30
|
Shah PT, Martin R, Yan Y, Shapiro JI, Liu J. Carbonylation Modification Regulates Na/K-ATPase Signaling and Salt Sensitivity: A Review and a Hypothesis. Front Physiol 2016; 7:256. [PMID: 27445847 PMCID: PMC4923243 DOI: 10.3389/fphys.2016.00256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/11/2016] [Indexed: 01/01/2023] Open
Abstract
Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions. Accumulating evidence indicates that oxidative stress not only regulates the Na/K-ATPase enzymatic activity, but also regulates its signaling and other functions. While cardiotonic steroids (CTS)-induced increase in reactive oxygen species (ROS) generation is an intermediate step in CTS-mediated Na/K-ATPase signaling, increase in ROS alone also stimulates Na/K-ATPase signaling. Based on literature and our observations, we hypothesize that ROS have biphasic effects on Na/K-ATPase signaling, transcellular sodium transport, and urinary sodium excretion. Oxidative modulation, in particular site specific carbonylation of the Na/K-ATPase α1 subunit, is a critical step in proximal tubular Na/K-ATPase signaling and decreased transcellular sodium transport leading to increases in urinary sodium excretion. However, once this system is overstimulated, the signaling, and associated changes in sodium excretion are blunted. This review aims to evaluate ROS-mediated carbonylation of the Na/K-ATPase, and its potential role in the regulation of pump signaling and sodium reabsorption in the renal proximal tubule (RPT).
Collapse
Affiliation(s)
- Preeya T Shah
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Rebecca Martin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Yanling Yan
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Joseph I Shapiro
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Jiang Liu
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| |
Collapse
|
31
|
Coughlan MT, Sharma K. Challenging the dogma of mitochondrial reactive oxygen species overproduction in diabetic kidney disease. Kidney Int 2016; 90:272-279. [PMID: 27217197 DOI: 10.1016/j.kint.2016.02.043] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/21/2016] [Accepted: 02/24/2016] [Indexed: 01/02/2023]
Abstract
The paradigm that high glucose drives overproduction of superoxide from mitochondria as a unifying theory to explain end organ damage in diabetes complications has been tightly held for more than a decade. With the recent development of techniques and probes to measure the production of distinct reactive oxygen species (ROS) in vivo, this widely held dogma is now being challenged with the emerging view that specific ROS moieties are essential for the function of specific intracellular signaling pathways and represent normal mitochondrial function. This review will provide a balanced overview of the dual nature of ROS, detailing current evidence for ROS overproduction in diabetic kidney disease, with a focus on cell types and sources of ROS. The technical aspects of measurement of mitochondrial ROS, both in isolated mitochondria and emerging in vivo methods will be discussed. The counterargument, that mitochondrial ROS production is reduced in diabetic complications, is consistent with a growing recognition that stimulation of mitochondrial biogenesis and oxidative phosphorylation activity reduces inflammation and fibrosis. It is clear that there is an urgent need to fully characterize ROS production paying particular attention to spatiotemporal aspects and to factor in the relevance of ROS in the regulation of cellular signaling in the pathogenesis of diabetic kidney disease. With improved tools and real-time imaging capacity, a greater understanding of the complex role of ROS will be able to guide novel therapeutic regimens.
Collapse
Affiliation(s)
- Melinda T Coughlan
- Baker International Diabetes Institute (IDI) Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Kumar Sharma
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Institute of Metabolomic Medicine, University of California-San Diego, La Jolla, California, USA; Division of Medical Genetics, Department of Medicine, University of California-San Diego, La Jolla, California, USA; Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, La Jolla, California, USA.
| |
Collapse
|
32
|
Song P, Onishi A, Koepsell H, Vallon V. Sodium glucose cotransporter SGLT1 as a therapeutic target in diabetes mellitus. Expert Opin Ther Targets 2016; 20:1109-25. [PMID: 26998950 DOI: 10.1517/14728222.2016.1168808] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Glycemic control is important in diabetes mellitus to minimize the progression of the disease and the risk of potentially devastating complications. Inhibition of the sodium-glucose cotransporter SGLT2 induces glucosuria and has been established as a new anti-hyperglycemic strategy. SGLT1 plays a distinct and complementing role to SGLT2 in glucose homeostasis and, therefore, SGLT1 inhibition may also have therapeutic potential. AREAS COVERED This review focuses on the physiology of SGLT1 in the small intestine and kidney and its pathophysiological role in diabetes. The therapeutic potential of SGLT1 inhibition, alone as well as in combination with SGLT2 inhibition, for anti-hyperglycemic therapy are discussed. Additionally, this review considers the effects on other SGLT1-expressing organs like the heart. EXPERT OPINION SGLT1 inhibition improves glucose homeostasis by reducing dietary glucose absorption in the intestine and by increasing the release of gastrointestinal incretins like glucagon-like peptide-1. SGLT1 inhibition has a small glucosuric effect in the normal kidney and this effect is increased in diabetes and during inhibition of SGLT2, which deliver more glucose to SGLT1 in late proximal tubule. In short-term studies, inhibition of SGLT1 and combined SGLT1/SGLT2 inhibition appeared to be safe. More data is needed on long-term safety and cardiovascular consequences of SGLT1 inhibition.
Collapse
Affiliation(s)
- Panai Song
- a Division of Nephrology & Hypertension, Department of Medicine , University of California San Diego , La Jolla , CA , USA.,b VA San Diego Healthcare System , San Diego , CA , USA.,c Department of Nephrology, Second Xiangya Hospital , Central South University , Changsha , China
| | - Akira Onishi
- a Division of Nephrology & Hypertension, Department of Medicine , University of California San Diego , La Jolla , CA , USA.,b VA San Diego Healthcare System , San Diego , CA , USA.,d Division of Nephrology, Department of Medicine , Jichi Medical University , Shimotsuke , Japan
| | - Hermann Koepsell
- e Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute , University of Würzburg , Würzburg , Germany
| | - Volker Vallon
- a Division of Nephrology & Hypertension, Department of Medicine , University of California San Diego , La Jolla , CA , USA.,b VA San Diego Healthcare System , San Diego , CA , USA.,f Department of Pharmacology , University of California San Diego , La Jolla , CA , USA
| |
Collapse
|
33
|
Slyvka Y, Malgor R, Inman SR, Ding J, Heh V, Nowak FV. Antioxidant diet and sex interact to regulate NOS isoform expression and glomerular mesangium proliferation in Zucker diabetic rat kidney. Acta Histochem 2016; 118:183-93. [PMID: 26797190 DOI: 10.1016/j.acthis.2015.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/30/2015] [Accepted: 12/31/2015] [Indexed: 12/21/2022]
Abstract
Oxidative stress contributes substantially to the pathophysiology of diabetic nephropathy (DN). Consumption of an antioxidant-fortified (AO) diet from an early age prevents or delays later development of DN in the Zucker rat female with type 2 diabetes. We hypothesize this is due to effects on mesangial matrix and renal nitric oxide synthase (NOS) distribution and to sex-specific differences in NOS responses in the diabetic kidney. Total glomerular tuft area (GTA) and PAS-positive tuft area (PTA), endothelial (e), neuronal (n) and inducible (i) NOS were quantified in males and females on AO or regular (REG) diet at 6 and 20 weeks of age. eNOS was observed in glomeruli and tubules. nNOS predominantly localized to tubular epithelium in both cortex and medulla. iNOS was expressed in proximal and distal tubules and collecting ducts. Sex, diabetes duration and AO diet affected the distribution of the three isoforms. GTA and PTA increased with duration of hyperglycemia and showed a negative correlation with renal levels of all NOS isoforms. AO diet in both genders was associated with less PAS-positive staining and less mesangial expansion than the REG diet, an early increase in cortical iNOS in males, and sex-specific changes in cortical eNOS at 20 weeks. These effects of AO diet may contribute to sex-specific preservation of renal function in females.
Collapse
Affiliation(s)
- Yuriy Slyvka
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA; The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA
| | - Ramiro Malgor
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA; Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA; The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA
| | - Sharon R Inman
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA; Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA; The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA
| | - Julia Ding
- College of Natural Sciences, School of Biological Sciences, University of Texas at Austin, TX 78713, USA
| | - Victor Heh
- OUHCOM Office of Research & Grants, Ohio University, Athens, OH 45701, USA
| | - Felicia V Nowak
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA; Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA; The Diabetes Institute at Ohio University, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
34
|
Ku SK, Bae JS. Vicenin-2 and scolymoside inhibit high-glucose-induced vascular inflammation in vitro and in vivo. Can J Physiol Pharmacol 2016; 94:287-95. [DOI: 10.1139/cjpp-2015-0215] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The vascular inflammatory process has been suggested to play a key role in the initiation and progression of atherosclerosis, a major complication of diabetes mellitus. Thus, in this study, we attempted to determine whether 2 structurally related flavonoids found in Cyclopia subternata, vicenin-2 and scolymoside, can suppress high-glucose (HG)-induced vascular inflammatory processes in human umbilical vein endothelial cells (HUVECs) and mice. The effects of vicenin-2 and scolymoside on HG-induced vascular inflammation were determined by measuring vascular permeability, leukocyte adhesion and migration, cell adhesion molecule (CAM) expression levels, and reactive oxygen species (ROS) formation. In addition, the anti-inflammation mechanism was investigated using immunofluorescence staining and Western blotting. The data showed that HG markedly increased vascular permeability, monocyte adhesion, expression of CAMs, formation of reactive oxygen species (ROS), and activation of nuclear factor (NF)-κB. Remarkably, pretreatment with vicenin-2 and scolymoside attenuated all of the above-mentioned vascular inflammatory effects of HG. HG-induced vascular inflammatory responses are critical events underlying the development of various diabetic complications; therefore, our results suggest that vicenin-2 and scolymoside have significant therapeutic benefits against diabetic complications and atherosclerosis.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 712-715, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea
| |
Collapse
|
35
|
Liu Z, Zhou Y, Yuan Y, Nie F, Peng R, Li Q, Lyu Z, Mao Z, Huang L, Zhou L, Li Y, Hao J, Ni D, Jin Q, Long Y, Ju P, Yu W, Liu J, Hu Y, Zhou Q. MiR542-3p Regulates the Epithelial-Mesenchymal Transition by Directly Targeting BMP7 in NRK52e. Int J Mol Sci 2015; 16:27945-55. [PMID: 26610487 PMCID: PMC4661932 DOI: 10.3390/ijms161126075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence demonstrated that miRNAs are highly involved in kidney fibrosis and Epithelial-Eesenchymal Transition (EMT), however, the mechanisms of miRNAs in kidney fibrosis are poorly understood. In this work, we identified that miR542-3p could promote EMT through down-regulating bone morphogenetic protein 7 (BMP7) expression by targeting BMP7 3′UTR. Firstly, real-time PCR results showed that miR542-3p was significantly up-regulated in kidney fibrosis in vitro and in vivo. Moreover, Western blot results demonstrated that miR542-3p may promote EMT in the NRK52e cell line. In addition, we confirmed that BMP7, which played a crucial role in anti-kidney fibrosis and suppressed the progression of EMT, was a target of miR542-3p through Dual-Luciferase reporter assay, as did Western blot analysis. The effects of miR542-3p on regulating EMT could also be suppressed by transiently overexpressing BMP7 in NRK52e cells. Taken together, miR542-3p may be a critical mediator of the induction of EMT via directly targeting BMP7.
Collapse
Affiliation(s)
- Zhicheng Liu
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Yuru Zhou
- The Seventh Class of 2012 year entry, the Third Clinical College, Chongqing Medical University, Chongqing 400016, China.
| | - Yue Yuan
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Fang Nie
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Rui Peng
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Qianyin Li
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Zhongshi Lyu
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Zhaomin Mao
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Liyuan Huang
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Li Zhou
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Yiman Li
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Jing Hao
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Dongsheng Ni
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Qianni Jin
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Yaoshui Long
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Pan Ju
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Wen Yu
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Jianing Liu
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Yanxia Hu
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Qin Zhou
- The Division of Molecular Nephrology and the Creative Training Center for Undergraduates, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
36
|
Subathra M, Korrapati M, Howell LA, Arthur JM, Shayman JA, Schnellmann RG, Siskind LJ. Kidney glycosphingolipids are elevated early in diabetic nephropathy and mediate hypertrophy of mesangial cells. Am J Physiol Renal Physiol 2015; 309:F204-15. [PMID: 26041445 DOI: 10.1152/ajprenal.00150.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022] Open
Abstract
Glycosphingolipids (GSLs) play a role in insulin resistance and diabetes, but their role in diabetic nephropathy (DN) has received limited attention. We used 9- and 17-wk-old nondiabetic db/m and diabetic db/db mice to examine the role of GSLs in DN. Cerebrosides or monoglycosylated GSLs [hexosylceramides (HexCers); glucosyl- and galactosylceramides] and lactosylceramide (LacCers) were elevated in db/db mouse kidney cortices, specifically in glomeruli, and also in urine. In our recent paper (25), we observed that the kidneys exhibited glomerular hypertrophy and proximal tubular vacuolization and increased fibrosis markers at these time points. Mesangial cells contribute to hyperglycemia-induced glomerular hypertrophy in DN. Hyperglycemic culture conditions, similar to that present in diabetes, were sufficient to elevate mesangial cell HexCers and increase markers of fibrosis, extracellular matrix proteins, and cellular hypertrophy. Inhibition of glucosylceramide synthase or lowering glucose levels decreased markers of fibrosis and extracellular matrix proteins and reversed mesangial cell hypertrophy. Hyperglycemia increased phosphorylated (p)SMAD3 and pAkt levels and reduced phosphatase and tensin homolog levels, which were reversed with glucosylceramide synthase inhibition. These data suggest that inhibition of glucosylceramide synthase reversed mesangial cell hypertrophy through decreased pAkt and pSmad3 and increased pathways responsible for protein degradation. Importantly, urinary GSL levels were higher in patients with DN compared with healthy control subjects, implicating a role for these lipids in human DN. Thus, hyperglycemia in type II diabetes leads to renal dysfunction at least in part by inducing accumulation of HexCers and LacCers in mesangial cells, resulting in fibrosis, extracellular matrix production, and hypertrophy.
Collapse
Affiliation(s)
- Marimuthu Subathra
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Midhun Korrapati
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren A Howell
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - John M Arthur
- University of Arkansas for Medical Sciences, Little Rock, Arkansas; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - James A Shayman
- Nephrology Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky;
| |
Collapse
|
37
|
Tojo A, Hatakeyama S, Kinugasa S, Nangaku M. Angiotensin receptor blocker telmisartan suppresses renal gluconeogenesis during starvation. Diabetes Metab Syndr Obes 2015; 8:103-13. [PMID: 25709483 PMCID: PMC4335621 DOI: 10.2147/dmso.s78771] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The kidney plays an important role in gluconeogenesis during starvation. To clarify the anti-diabetic action of angiotensin receptor blockers, we examined the effects of telmisartan on the sodium-glucose co-transporters (SGLT) and the pathways of renal gluconeogenesis in streptozotocin-induced diabetes mellitus (DM) rats. At 4 weeks, the DM rats treated with/without telmisartan for 2 weeks and normal control rats were used for the study after a 24-hour fast. SGLT2 expressed on the brush border membrane of the proximal convoluted tubules increased in the DM rats, but decreased in the rats treated with telmisartan. The expression of restriction enzymes of gluconeogenesis, glucose-6-phosphatase, and phosphoenolpyruvate carboxykinase increased in the proximal tubules in the DM rats, whereas these enzymes decreased in the kidneys of the rats treated with telmisartan. The elevated cytoplasmic glucose-6-phosphate and glucose levels in the kidney of DM rats significantly decreased in those treated with telmisartan, whereas those levels in the liver did not show significant change. Meanwhile, the high plasma glucose levels in the DM rats during the intravenous insulin tolerance tests were ameliorated by telmisartan. The increased fasting plasma glucose levels after 24 hours of starvation in the DM rats thus returned to the control levels by telmisartan treatment. In conclusion, the increased renal SGLT2 expression, elevated renal gluconeogenesis enzymes and extent of insulin-resistance in the DM rats were ameliorated by telmisartan therapy, thus resulting in decreased plasma glucose levels after 24 hours of fasting.
Collapse
Affiliation(s)
- Akihiro Tojo
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
- Correspondence: Akihiro Tojo, Division of Nephrology and Endocrinology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan, Tel +81 3 3815 5411 ext 37219, Fax +81 3 3814 0021, Email
| | - Saaya Hatakeyama
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Satoshi Kinugasa
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
Chao CT, Chiang CK. Uremic toxins, oxidative stress, and renal fibrosis: an interwined complex. J Ren Nutr 2014; 25:155-9. [PMID: 25511523 DOI: 10.1053/j.jrn.2014.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/29/2014] [Indexed: 11/11/2022] Open
Abstract
The prevalence of end-stage renal diseases is currently on the rise globally, and finding the way to curb this tide is urgently needed. Tubulointerstitial fibrosis is a common pathway for essentially all the nephropathy categories known to date, and the manifestations of renal fibrosis include excessive deposition of extracellular matrix with distortion of renal microstructures and functional deterioration. Uremic toxins have been gradually found to play an important role in the development of progressive renal fibrosis, with protein-bound indoxyl sulfate, p-cresol, and p-cresyl sulfate receiving the most attention. However, the contribution of oxidative stress among the pathogenesis of uremic toxins and renal fibrosis has not been evaluated much until recently. In this review, we will discuss about the nature and sources of oxidative stress in the kidney and how uremic toxins use oxidative stress to orchestrate the processes of renal fibrosis.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Kang Chiang
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Integrative Diagnostics and Therapeutics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
39
|
Lee W, Ku SK, Lee D, Lee T, Bae JS. Emodin-6-O-β-D--glucoside inhibits high-glucose-induced vascular inflammation. Inflammation 2014; 37:306-13. [PMID: 24114447 DOI: 10.1007/s10753-013-9741-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Emodin-6-O-β-D-glucoside (EG), a new active compound from Reynoutria japonica, has recently been shown to exert potent anti-inflammatory and barrier protective effects in human umbilical vein endothelial cells (HUVECs) and in mice. Vascular inflammatory process has been suggested to play a key role in initiation and progression of atherosclerosis, a major complication of diabetes mellitus. Thus, we attempted to determine whether EG can suppress the vascular inflammatory process induced by high glucose (HG) in HUVECs and mice. Data showed that HG induced markedly increased vascular permeability, monocyte adhesion, expressions of CAMs, formation of ROS, and activation of NF-κB. Remarkably, all of the above-mentioned vascular inflammatory effects of HG were attenuated by pretreatment with EG. Vascular inflammatory responses induced by HG are critical events underlying development of various diabetic complications; therefore, our results suggest that EG may have significant therapeutic benefits against diabetic complications and atherosclerosis.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | | | | | | | | |
Collapse
|
40
|
Aspalathin and Nothofagin from Rooibos (Aspalathus linearis) Inhibits High Glucose-Induced Inflammation In Vitro and In Vivo. Inflammation 2014; 38:445-55. [DOI: 10.1007/s10753-014-0049-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Sun T, Yang J, Dong W, Wang R, Ma P, Kang P, Zhang H, Xie C, Du J, Zhao L. Down-regulated miR-15a mediates the epithelial–mesenchymal transition in renal tubular epithelial cells promoted by high glucose. Biosci Biotechnol Biochem 2014; 78:1363-70. [PMID: 25130738 DOI: 10.1080/09168451.2014.936345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Abstract
High glucose (HG) has been reported to be associated with renal dysfunction. And one potential mechanism underlining the dysfunction is the epithelial–mesenchymal transition (EMT) of renal tubular epithelial cells. Present study showed that EMT was induced in the HG-treated renal tubular epithelial cells by promoting the expression of mesenchymal phenotype molecules, such as α-SMA and collagen I, and down-regulating the expression of epithelial phenotype molecule E-cadherin. Moreover, we have identified the down-regulation of miR-15a which was accompanied with the HG-induced EMT. And the miR-15a overexpression inhibited the α-SMA, collagen I expression, and the promotion of E-cadherin expression by targeting and down-regulating AP4 which was also significantly promoted by the HG in the renal tubular epithelial cells. Thus, this study revealed that the weakening regulation on the AP4 expression by miR-15a might contribute to the HG-induced EMT in the renal tubular epithelial cells.
Collapse
Affiliation(s)
- Tingli Sun
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Jun Yang
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Wenpeng Dong
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Ruiyan Wang
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Peilong Ma
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Ping Kang
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Hongbo Zhang
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Changying Xie
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Juan Du
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, China
| | - Lijie Zhao
- Department of Geriatrics, General Hospital of Daqing Oil Field, Daqing, China
| |
Collapse
|
42
|
Hsu YC, Lee PH, Lei CC, Ho C, Shih YH, Lin CL. Nitric oxide donors rescue diabetic nephropathy through oxidative-stress-and nitrosative-stress-mediated Wnt signaling pathways. J Diabetes Investig 2014; 6:24-34. [PMID: 25621130 PMCID: PMC4296700 DOI: 10.1111/jdi.12244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 03/14/2014] [Accepted: 04/06/2014] [Indexed: 12/13/2022] Open
Abstract
AIMS/INTRODUCTION The role of the renal nitric oxide (NO) system in the pathophysiology of diabetic nephropathy constitutes a very challenging and fertile field for future investigation. The purpose of the present study was to investigate whether NO donors can attenuate diabetic renal fibrosis and apoptosis through modulating oxidative-and nitrosative-stress, and Wnt signaling using in vivo diabetic models. MATERIALS AND METHODS Diabetic rat was induced by a single intraperitoneal injection of streptozotocin. Rats in each group were intraperitoneally given 2,2'-(hydroxynitrosohydrazino)bis-ethanamine (1 U/kg/day) and vehicle for 28 and 56 consecutive days. Expression of the oxidative-and nitrosative-stress, and Wnt signaling components were examined in kidneys from diabetic animals by quantitative reverse transcription polymerase chain reaction, western blot analysis and immunohistochemical staining. RESULTS NO donor treatment significantly reduced the ratio of kidney weight to bodyweight and proteinuria. This treatment also significantly restored the suppressive effect of diabetes on urinary NO2 + NO3 levels. Immunohistochemistry showed that NO donor treatment significantly reduced transforming growth factor (TGF)-β1, fibronectin, cleaved caspase-3 and triphosphate-biotin nick end-labeling expression in the glomeruli of diabetic rats. We found that diabetes promoted 8-hydroxy-2'-deoxyguanosine, and peroxynitrite expression coincided with reduced endothelial NO synthase expression in glomeruli. Interestingly, NO donor treatment completely removed oxidative stress and nitrosative stress, and restored endothelial NO synthase expression in diabetic renal glomeruli. Immunohistomorphometry results showed that NO donor treatment significantly restored suppressed Wnt5a expression and β-catenin immunoreactivities in glomeruli. Based on laser-captured microdissection for quantitative reverse transcription polymerase chain reaction, diabetes significantly increased TGF-β1, and fibronectin expression coincided with depressed Wnt5a expression. NO donor treatment reduced TGF-β1, fibronectin activation, and the suppressing effect of diabetes on Wnt5a and β-catenin expression in renal glomeruli. CONCLUSIONS NO donor treatment alleviates extracellular matrix accumulation and apoptosis in diabetic nephropathy in vivo by not only preventing the diabetes-mediated oxidative and nitrostative stress, but also restoring downregulation of endothelial NO synthase expression and Wnt/β-catenin signaling. These findings suggest that modulation of NO is a viable alternative strategy for rescuing diabetic renal injury.
Collapse
Affiliation(s)
- Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital Chiayi, Taiwan ; Department of Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital Chiayi, Taiwan ; Chronic Kidney Disease Center, Chang Gung Memorial Hospital Chiayi, Taiwan ; Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital Chiayi, Taiwan
| | - Pei-Hsien Lee
- Department of Nephrology, Chang Gung Memorial Hospital Chiayi, Taiwan ; Department of Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital Chiayi, Taiwan ; Chronic Kidney Disease Center, Chang Gung Memorial Hospital Chiayi, Taiwan ; Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital Chiayi, Taiwan
| | - Chen-Chou Lei
- Department of Nephrology, Chang Gung Memorial Hospital Chiayi, Taiwan ; Department of Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital Chiayi, Taiwan ; Chronic Kidney Disease Center, Chang Gung Memorial Hospital Chiayi, Taiwan ; Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital Chiayi, Taiwan
| | - Cheng Ho
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital Chiayi, Taiwan
| | - Ya-Hsueh Shih
- Department of Nephrology, Chang Gung Memorial Hospital Chiayi, Taiwan ; Department of Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital Chiayi, Taiwan ; Chronic Kidney Disease Center, Chang Gung Memorial Hospital Chiayi, Taiwan
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital Chiayi, Taiwan ; Department of Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital Chiayi, Taiwan ; Chronic Kidney Disease Center, Chang Gung Memorial Hospital Chiayi, Taiwan ; School of Traditional Chinese Medicine, Chang Gung University College of Medicine Tao-Yuan, Taiwan
| |
Collapse
|
43
|
Orientin Inhibits High Glucose-Induced Vascular Inflammation In Vitro and In Vivo. Inflammation 2014; 37:2164-73. [DOI: 10.1007/s10753-014-9950-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
44
|
Kwak S, Ku SK, Bae JS. Fisetin inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflamm Res 2014; 63:779-87. [PMID: 24923846 DOI: 10.1007/s00011-014-0750-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/08/2014] [Accepted: 05/30/2014] [Indexed: 11/28/2022] Open
Abstract
AIM AND OBJECTIVE Fisetin, an active compound isolated from flowering plants in the family Fabaceae, was reported to have antiviral, neuroprotective, and anti-inflammatory effects. Vascular inflammatory processes have been suggested to play key roles in the initiation and progression of atherosclerosis, a major complication of diabetes mellitus. Thus, we determined the ability of fisetin to suppress vascular inflammatory processes induced by high glucose (HG) in primary human umbilical vein endothelial cells (HUVECs) and mice. METHODS The effects of fisetin on HG-induced vascular inflammation were determined by measuring vascular permeability, leukocyte adhesion and migration, cell adhesion molecule (CAM) expression levels, reactive oxygen species (ROS) formation, and nuclear factor (NF)-κB activation. RESULTS HG markedly increased vascular permeability, monocyte adhesion, expressions of CAMs, formation of ROS, and activation of NF-κB. Remarkably, all of the observed vascular inflammatory effects induced by HG were inhibited by pretreatment with fisetin. CONCLUSION Vascular inflammatory responses induced by HG are critical events underlying the development of diabetic complications; therefore, our results suggest that fisetin possesses significant therapeutic effects against diabetic complications and atherosclerosis.
Collapse
Affiliation(s)
- Soyoung Kwak
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro Buk-gu, Daegu, 702-701, Republic of Korea
| | | | | |
Collapse
|
45
|
De Nicola L, Gabbai FB, Liberti ME, Sagliocca A, Conte G, Minutolo R. Sodium/glucose cotransporter 2 inhibitors and prevention of diabetic nephropathy: targeting the renal tubule in diabetes. Am J Kidney Dis 2014; 64:16-24. [PMID: 24673844 DOI: 10.1053/j.ajkd.2014.02.010] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/05/2014] [Indexed: 12/25/2022]
Abstract
Optimal prevention and treatment of chronic kidney disease in diabetes requires implementing therapies that specifically interfere with the pathogenesis of diabetic nephropathy. In this regard, significant attention has been given to alterations of the proximal tubule and resulting changes in glomerular filtration rate. At the onset of diabetes mellitus, hyperglycemia causes increases in proximal tubular reabsorption secondary to induction of tubular growth with associated increases in sodium/glucose cotransport. The increase in proximal reabsorption leads to a decrease in solute load to the macula densa, deactivation of the tubuloglomerular feedback, and increases in glomerular filtration rate. Because glomerular hyperfiltration currently is recognized as a risk factor for progression of kidney disease in diabetic patients, limiting proximal tubular reabsorption constitutes a potential target to reduce hyperfiltration. The recent introduction of sodium/glucose cotransporter 2 (SGLT2) inhibitors opens new therapeutic perspectives for this high-risk patient population. Experimental studies have shown that these new agents attenuate the progressive nature of diabetic nephropathy by blood glucose-dependent and -independent mechanisms. SGLT2 inhibition may prevent glomerular hyperfiltration independent of the effect of lowering blood glucose levels while limiting kidney growth, inflammation, and albuminuria through reductions in blood glucose levels. Clinical data for the potential role of the proximal tubule in the pathophysiology of diabetic nephropathy and the nephroprotective effects of SGLT2 inhibitors currently are limited compared to the more extensive experimental literature. We review the evidence supporting this working hypothesis by integrating the experimental findings with the available clinical data.
Collapse
Affiliation(s)
- Luca De Nicola
- Nephrology Division, Second University of Naples-Med School, Naples, Italy.
| | - Francis B Gabbai
- Department of Medicine, Veterans Administration San Diego Healthcare System-University of California at San Diego Medical School, San Diego, CA
| | | | - Adelia Sagliocca
- Nephrology Division, Second University of Naples-Med School, Naples, Italy
| | - Giuseppe Conte
- Nephrology Division, Second University of Naples-Med School, Naples, Italy
| | - Roberto Minutolo
- Nephrology Division, Second University of Naples-Med School, Naples, Italy
| |
Collapse
|
46
|
Ku SK, Kwak S, Kwon OJ, Bae JS. Hyperoside Inhibits High-Glucose-Induced Vascular Inflammation In Vitro and In Vivo. Inflammation 2014; 37:1389-400. [DOI: 10.1007/s10753-014-9863-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
47
|
Giacco F, Du X, D’Agati VD, Milne R, Sui G, Geoffrion M, Brownlee M. Knockdown of glyoxalase 1 mimics diabetic nephropathy in nondiabetic mice. Diabetes 2014; 63:291-9. [PMID: 24062246 PMCID: PMC3868051 DOI: 10.2337/db13-0316] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Differences in susceptibility to diabetic nephropathy (DN) between mouse strains with identical levels of hyperglycemia correlate with renal levels of oxidative stress, shown previously to play a central role in the pathogenesis of DN. Susceptibility to DN appears to be genetically determined, but the critical genes have not yet been identified. Overexpression of the enzyme glyoxalase 1 (Glo1), which prevents posttranslational modification of proteins by the glycolysis-derived α-oxoaldehyde, methylglyoxal (MG), prevents hyperglycemia-induced oxidative stress in cultured cells and model organisms. In this study, we show that in nondiabetic mice, knockdown of Glo1 increases to diabetic levels both MG modification of glomerular proteins and oxidative stress, causing alterations in kidney morphology indistinguishable from those caused by diabetes. We also show that in diabetic mice, Glo1 overexpression completely prevents diabetes-induced increases in MG modification of glomerular proteins, increased oxidative stress, and the development of diabetic kidney pathology, despite unchanged levels of diabetic hyperglycemia. Together, these data indicate that Glo1 activity regulates the sensitivity of the kidney to hyperglycemic-induced renal pathology and that alterations in the rate of MG detoxification are sufficient to determine the glycemic set point at which DN occurs.
Collapse
Affiliation(s)
- Ferdinando Giacco
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Xueliang Du
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Vivette D. D’Agati
- Division of Renal Pathology, Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Ross Milne
- Diabetes and Atherosclerosis Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Guangzhi Sui
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Michele Geoffrion
- Diabetes and Atherosclerosis Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Michael Brownlee
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Corresponding author: Michael Brownlee,
| |
Collapse
|
48
|
Tchounwou CK, Yedjou CG, Farah I, Tchounwou PB. D-Glucose-Induced Cytotoxic, Genotoxic, and Apoptotic Effects on Human Breast Adenocarcinoma (MCF-7) Cells. ACTA ACUST UNITED AC 2014; 6:156-160. [PMID: 25506409 PMCID: PMC4264661 DOI: 10.4172/1948-5956.1000265] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction Glucose is a simple sugar that plays an important role in energy production in biological systems. However, it has been linked to many long-term health problems including the risk of heart disease and stroke, erectile dysfunction in men and pregnancy complications in women, and damage to the kidneys, nerves, eye and vision. Also, the underlying mechanisms of diabetic complications are poorly understood. Methods In the present study, D-glucose-induced cytotoxic, genotoxic, and apoptotic effects were studied using MCF-7 cells as an in vitro test model. Cell viability was determined by MTT assay. Genotoxic damage was tested by the means of alkaline single cell gel electrophoresis (Comet) assay. Cell apoptosis was measured by flow cytometry assessment (Annexin-V/PI assay). Results The results of MTT assay indicated that D-glucose significantly reduces the viability of MCF-7 cells in a dose and time-dependent manner. Similar trend was obtained with the trypan blue exclusion test. Data obtained from the Comet assay indicated that D-glucose causes DNA damage in MCF-7 cells in a dose-dependent manner. The flow cytometry assessment (Annexin V FITC/PI) showed a strong dose-response relationship between D-glucose exposure and annexin V positive MCF-7 cells undergoing early apoptosis. Conclusion Taking together, these data provide clear evidence that D-glucose induces cytotoxic, genotoxic, and apoptotic effects on MCF-7 cells. This finding represents the basis for further studies addressing the pathophysiological mechanisms of action of glucose overdose.
Collapse
Affiliation(s)
- Christine K Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, USA
| | - Clement G Yedjou
- Cellomics and Toxicogenomics Research Laboratory, NIH-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, USA
| | - Ibrahim Farah
- Cellomics and Toxicogenomics Research Laboratory, NIH-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, USA
| | - Paul B Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, USA
| |
Collapse
|
49
|
A glimpse of the pathogenetic mechanisms of Wnt/β-catenin signaling in diabetic nephropathy. BIOMED RESEARCH INTERNATIONAL 2013; 2013:987064. [PMID: 24455745 PMCID: PMC3886614 DOI: 10.1155/2013/987064] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/22/2013] [Indexed: 01/22/2023]
Abstract
The Wnt family of proteins belongs to a group of secreted lipid-modified glycoproteins with highly conserved cysteine residues. Prior results indicate that Wnt/β-catenin signaling plays a prominent role in cell differentiation, adhesion, survival, and apoptosis and is involved in organ development, tumorigenesis, and tissue fibrosis, among other functions. Accumulating evidence has suggested that Wnt/β-catenin exhibits a pivotal function in the progression of diabetic nephropathy (DN). In this review, we focused on discussing the dual role of Wnt/β-catenin in apoptosis and epithelial mesenchymal transition (EMT) formation of mesangial cells. Moreover, we also elucidated the effect of Wnt/β-catenin in podocyte dysfunction, tubular EMT formation, and renal fibrosis under DN conditions. In addition, the molecular mechanisms involved in this process are introduced. This information provides a novel molecular target of Wnt/β-catenin for the protection of kidney damage and in delay of the progression of DN.
Collapse
|
50
|
Abstract
Diabetes mellitus contributes greatly to morbidity, mortality, and overall health care costs. In major part, these outcomes derive from the high incidence of progressive kidney dysfunction in patients with diabetes making diabetic nephropathy a leading cause of end-stage renal disease. A better understanding of the molecular mechanism involved and of the early dysfunctions observed in the diabetic kidney may permit the development of new strategies to prevent diabetic nephropathy. Here we review the pathophysiological changes that occur in the kidney in response to hyperglycemia, including the cellular responses to high glucose and the responses in vascular, glomerular, podocyte, and tubular function. The molecular basis, characteristics, and consequences of the unique growth phenotypes observed in the diabetic kidney, including glomerular structures and tubular segments, are outlined. We delineate mechanisms of early diabetic glomerular hyperfiltration including primary vascular events as well as the primary role of tubular growth, hyperreabsorption, and tubuloglomerular communication as part of a "tubulocentric" concept of early diabetic kidney function. The latter also explains the "salt paradox" of the early diabetic kidney, that is, a unique and inverse relationship between glomerular filtration rate and dietary salt intake. The mechanisms and consequences of the intrarenal activation of the renin-angiotensin system and of diabetes-induced tubular glycogen accumulation are discussed. Moreover, we aim to link the changes that occur early in the diabetic kidney including the growth phenotype, oxidative stress, hypoxia, and formation of advanced glycation end products to mechanisms involved in progressive kidney disease.
Collapse
Affiliation(s)
- Volker Vallon
- Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA.
| | | |
Collapse
|