1
|
Downie ML, Desjarlais A, Verdin N, Woodlock T, Collister D. Precision Medicine in Diabetic Kidney Disease: A Narrative Review Framed by Lived Experience. Can J Kidney Health Dis 2023; 10:20543581231209012. [PMID: 37920777 PMCID: PMC10619345 DOI: 10.1177/20543581231209012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/10/2023] [Indexed: 11/04/2023] Open
Abstract
Purpose of review Diabetic kidney disease (DKD) is a leading cause of chronic kidney disease (CKD) for which many treatments exist that have been shown to prevent CKD progression and kidney failure. However, DKD is a complex and heterogeneous etiology of CKD with a spectrum of phenotypes and disease trajectories. In this narrative review, we discuss precision medicine approaches to DKD, including genomics, metabolomics, proteomics, and their potential role in the management of diabetes mellitus and DKD. A patient and caregivers of patients with lived experience with CKD were involved in this review. Sources of information Original research articles were identified from MEDLINE and Google Scholar using the search terms "diabetes," "diabetic kidney disease," "diabetic nephropathy," "chronic kidney disease," "kidney failure," "dialysis," "nephrology," "genomics," "metabolomics," and "proteomics." Methods A focused review and critical appraisal of existing literature regarding the precision medicine approaches to the diagnosis, prognosis, and treatment of diabetes and DKD framed by a patient partner's/caregiver's lived experience. Key findings Distinguishing diabetic nephropathy from CKD due to other types of DKD and non-DKD is challenging and typically requires a kidney biopsy for a diagnosis. Biomarkers have been identified to assist with the prediction of the onset and progression of DKD, but they have yet to be incorporated and evaluated relative to clinical standard of care CKD and kidney failure risk prediction tools. Genomics has identified multiple causal genetic variants for neonatal diabetes mellitus and monogenic diabetes of the young that can be used for diagnostic purposes and to specify antiglycemic therapy. Genome-wide-associated studies have identified genes implicated in DKD pathophysiology in the setting of type 1 and 2 diabetes but their translational benefits are lagging beyond polygenetic risk scores. Metabolomics and proteomics have been shown to improve diagnostic accuracy in DKD, have been used to identify novel pathways involved in DKD pathogenesis, and can be used to improve the prediction of CKD progression and kidney failure as well as predict response to DKD therapy. Limitations There are a limited number of large, high-quality prospective observational studies and no randomized controlled trials that support the use of precision medicine based approaches to improve clinical outcomes in adults with or at risk of diabetes and DKD. It is unclear which patients may benefit from the clinical use of genomics, metabolomics and proteomics along the spectrum of DKD trajectory. Implications Additional research is needed to evaluate the role of the use of precision medicine for DKD management, including diagnosis, differentiation of diabetic nephropathy from other etiologies of DKD and CKD, short-term and long-term risk prognostication kidney outcomes, and the prediction of response to and safety of disease-modifying therapies.
Collapse
Affiliation(s)
- Mallory L. Downie
- McGill University Health Center Research Institute, Montreal, QC, Canada
| | - Arlene Desjarlais
- Kidney Research Scientist Core Education and National Training Program, Montreal, QC, Canada
| | - Nancy Verdin
- Kidney Research Scientist Core Education and National Training Program, Montreal, QC, Canada
| | - Tania Woodlock
- Kidney Research Scientist Core Education and National Training Program, Montreal, QC, Canada
| | - David Collister
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
2
|
Lassén E, Daehn IS. Molecular Mechanisms in Early Diabetic Kidney Disease: Glomerular Endothelial Cell Dysfunction. Int J Mol Sci 2020; 21:ijms21249456. [PMID: 33322614 PMCID: PMC7764016 DOI: 10.3390/ijms21249456] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD), with prevalence increasing at an alarming rate worldwide and today, there are no known cures. The pathogenesis of DKD is complex, influenced by genetics and the environment. However, the underlying molecular mechanisms that contribute to DKD risk in about one-third of diabetics are still poorly understood. The early stage of DKD is characterized by glomerular hyperfiltration, hypertrophy, podocyte injury and depletion. Recent evidence of glomerular endothelial cell injury at the early stage of DKD has been suggested to be critical in the pathological process and has highlighted the importance of glomerular intercellular crosstalk. A potential mechanism may include reactive oxygen species (ROS), which play a direct role in diabetes and its complications. In this review, we discuss different cellular sources of ROS in diabetes and a new emerging paradigm of endothelial cell dysfunction as a key event in the pathogenesis of DKD.
Collapse
|
3
|
Satari M, Aghadavod E, Mobini M, Asemi Z. Association between miRNAs expression and signaling pathways of oxidative stress in diabetic retinopathy. J Cell Physiol 2018; 234:8522-8532. [PMID: 30478922 DOI: 10.1002/jcp.27801] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/31/2018] [Indexed: 12/12/2022]
Abstract
Diabetic retinopathy (DR) is a major cause of vision reduction in diabetic patients. Hyperglycemia is a known instigator for the development of DR, even though the role of oxidative stress pathways in the pathogenesis of DR is established. The studies indicate that microRNAs (miRNAs) are significant to the etiology of DR; changes in miRNAs expression levels may be associated with onset and progression of DR. In addition, miRNAs have emerged as a useful disease marker due to their availability and stability in detecting the severity of DR. The relationship between miRNAs expression levels and oxidative stress pathways has been investigated in several studies. The aim of this study is the examination of function and expression levels of target miRNAs in oxidative stress pathway and pathogenesis of diabetic retinopathy.
Collapse
Affiliation(s)
- Mahbobeh Satari
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Depatrment of Biochemistry, Kashan University of Medical Sciences, Kashan, Iran
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Depatrment of Biochemistry, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Mobini
- Department of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Depatrment of Biochemistry, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
4
|
Mahavadi S, Sriwai W, Manion O, Grider JR, Murthy KS. Diabetes-induced oxidative stress mediates upregulation of RhoA/Rho kinase pathway and hypercontractility of gastric smooth muscle. PLoS One 2017; 12:e0178574. [PMID: 28678840 PMCID: PMC5497948 DOI: 10.1371/journal.pone.0178574] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of diabetes-associated motility disorders are multifactorial and attributed to abnormalities in extrinsic and intrinsic innervation, and a decrease in the number of interstitial cells of Cajal, and nNOS expression and activity. Here we studied the effect of hyperglycemia on smooth muscle function. Using smooth muscles from the fundus of ob/ob mice and of wild type (WT) mice treated with 30 mM glucose (HG), we identified the molecular mechanism by which hyperglycemia upregulates RhoA/Rho kinase pathway and muscle contraction. RhoA expression, Rho kinase activity and muscle contraction were increased, while miR-133a expression was decreased in smooth muscle of ob/ob mice and in smooth muscle treated with HG. Intraperitoneal injections of pre-miR-133a decreased RhoA expression in WT mice and reversed the increase in RhoA expression in ob/ob mice. Intraperitoneal injections of antagomiR-133a increased RhoA expression in WT mice and augmented the increase in RhoA expression in ob/ob mice. The effect of pre-miR-133a or antagomiR-133a in vitro in smooth muscle treated with HG was similar to that obtained in vivo, suggesting that the expression of RhoA is negatively regulated by miR-133a and a decrease in miR-133a expression in diabetes causes an increase in RhoA expression. Oxidative stress (levels of reactive oxygen species and hydrogen peroxide, and expression of superoxide dismutase 1 and NADPH oxidase 4) was increased in smooth muscle of ob/ob mice and in HG-treated smooth muscle. Treatment of ob/ob mice with N-acetylcysteine (NAC) in vivo or addition of NAC in vitro to HG-treated smooth muscle reversed the effect of glucose on the expression of miR-133a and RhoA, Rho kinase activity and muscle contraction. NAC treatment also reversed the decrease in gastric emptying in ob/ob mice. We conclude that oxidative stress in diabetes causes a decrease in miR-133a expression leading to an increase in RhoA/Rho kinase pathway and muscle contraction.
Collapse
Affiliation(s)
- Sunila Mahavadi
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Wimolpak Sriwai
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Olivia Manion
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - John R. Grider
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Karnam S. Murthy
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
5
|
Rayegan S, Dehpour AR, Sharifi AM. Studying neuroprotective effect of Atorvastatin as a small molecule drug on high glucose-induced neurotoxicity in undifferentiated PC12 cells: role of NADPH oxidase. Metab Brain Dis 2017; 32:41-49. [PMID: 27476541 PMCID: PMC7102122 DOI: 10.1007/s11011-016-9883-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 07/25/2016] [Indexed: 01/01/2023]
Abstract
Overproduction of reactive oxygen species (ROS) by NADPH oxidase (NOX) activation has been considered the essential mechanism induced by hyperglycemia in various tissues. However, there is no comprehensive study on the role of NOXs in high glucose (HG)-induced toxic effect in neural tissues. Recently, a therapeutic strategy in oxidative related pathologies has been introduced by blocking the undesirable actions of NOX enzymes by small molecules. The protective roles of Statins in ameliorating oxidative stress by NOX inhibition have been shown in some tissues except neural. We hypothesized then, that different NOXs may have role in HG-induced neural cell injury. Furthermore, we postulate that Atorvastatin as a small molecule may modulate this NOXs activity to protect neural cells. Undifferentiated PC12 cells were treated with HG (140 mM/24 h) in the presence and absence of Atorvastatin (1 μM/96 h). The cell viability was measured by MTT assay and the gene and protein expressions profile of NOX (1-4) were determined by RT-PCR and western blotting, respectively. Levels of ROS and malondialdehyde (MDA) were also evaluated. Gene and protein expression levels of NOX (1-4) and consequently ROS and MDA levels were elevated in HG-treated PC12 cells. Atorvastatin could significantly decrease HG-induced NOXs, ROS and MDA elevation and improve impaired cell viability. It can be concluded that HG could elevate NOXs activity, ROS and MDA levels in neural tissues and Atorvastatin as a small molecule NOX inhibitor drug may prevent and delay diabetic complications, particularly neuropathy.
Collapse
Affiliation(s)
- Samira Rayegan
- Razi Drug Research Center and Dept. of Pharmacology, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Sharifi
- Razi Drug Research Center and Dept. of Pharmacology, Iran University of Medical Sciences, Tehran, Iran.
- Department of Tissue engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Tissue engineering group, Department of Orthopedics surgery, Faculty of Medicine, University of Malaya, Kuala lumpur, Malaysia.
| |
Collapse
|
6
|
Ma R, Chaudhari S, Li W. Canonical Transient Receptor Potential 6 Channel: A New Target of Reactive Oxygen Species in Renal Physiology and Pathology. Antioxid Redox Signal 2016; 25:732-748. [PMID: 26937558 PMCID: PMC5079416 DOI: 10.1089/ars.2016.6661] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/06/2016] [Indexed: 02/07/2023]
Abstract
SIGNIFICANCE Regulation of Ca2+ signaling cascade by reactive oxygen species (ROS) is becoming increasingly evident and this regulation represents a key mechanism for control of many fundamental cellular functions. Canonical transient receptor potential (TRPC) 6, a member of Ca2+-conductive channel in the TRPC family, is widely expressed in kidney cells, including glomerular mesangial cells, podocytes, tubular epithelial cells, and vascular myocytes in renal microvasculature. Both overproduction of ROS and dysfunction of TRPC6 channel are involved in renal injury in animal models and human subjects. Although regulation of TRPC channel function by ROS has been well described in other tissues and cell types, such as vascular smooth muscle, this important cell regulatory mechanism has not been fully reviewed in kidney cells. Recent Advances: Accumulating evidence has shown that TRPC6 is a redox-sensitive channel, and modulation of TRPC6 Ca2+ signaling by altering TRPC6 protein expression or TRPC6 channel activity in kidney cells is a downstream mechanism by which ROS induce renal damage. CRITICAL ISSUES This review highlights how recent studies analyzing function and expression of TRPC6 channels in the kidney and their response to ROS improve our mechanistic understanding of oxidative stress-related kidney diseases. FUTURE DIRECTIONS Although it is evident that ROS regulate TRPC6-mediated Ca2+ signaling in several types of kidney cells, further study is needed to identify the underlying molecular mechanism. We hope that the newly identified ROS/TRPC6 pathway will pave the way to new, promising therapeutic strategies to target kidney diseases such as diabetic nephropathy. Antioxid. Redox Signal. 25, 732-748.
Collapse
Affiliation(s)
- Rong Ma
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas
| | - Sarika Chaudhari
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas
| | - Weizu Li
- Department of Pharmacology, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
7
|
Quercetin-Rich Guava (Psidium guajava) Juice in Combination with Trehalose Reduces Autophagy, Apoptosis and Pyroptosis Formation in the Kidney and Pancreas of Type II Diabetic Rats. Molecules 2016; 21:334. [PMID: 26978332 PMCID: PMC6274302 DOI: 10.3390/molecules21030334] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/28/2016] [Accepted: 03/02/2016] [Indexed: 02/07/2023] Open
Abstract
We explored whether the combination of anti-oxidant and anti-inflammatory guava (Psidium guajava) and trehalose treatment protects the kidney and pancreas against Type II diabetes (T2DM)-induced injury in rats. We measured the active component of guava juice by HPLC analysis. T2DM was induced in Wistar rats by intraperitoneal administration of nicotinamide and streptozotocin and combination with high fructose diets for 8 weeks. The rats fed with different dosages of guava juice in combination with or without trehalose for 4 weeks were evaluated the parameters including OGTT, plasma insulin, HbA1c, HOMA-IR (insulin resistance) and HOMA-β (β cell function and insulin secretion). We measured oxidative and inflammatory degrees by immunohistochemistry stain, fluorescent stain, and western blot and serum and kidney reactive oxygen species (ROS) by a chemiluminescence analyzer. High content of quercetin in the guava juice scavenged H2O2 and HOCl, whereas trehalose selectively reduced H2O2, not HOCl. T2DM affected the levels in OGTT, plasma insulin, HbA1c, HOMA-IR and HOMA-β, whereas these T2DM-altered parameters, except HbA1c, were significantly improved by guava and trehalose treatment. The levels of T2DM-enhanced renal ROS, 4-hydroxynonenal, caspase-3/apoptosis, LC3-B/autophagy and IL-1β/pyroptosis were significantly decreased by guava juice and trehalose. The combination with trehalose and guava juice protects the pancreas and kidney against T2DM-induced injury.
Collapse
|
8
|
Koulis C, Watson A, Gray S, Jandeleit-Dahm K. Linking RAGE and Nox in diabetic micro- and macrovascular complications. DIABETES & METABOLISM 2015; 41:272-281. [DOI: 10.1016/j.diabet.2015.01.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 01/24/2015] [Accepted: 01/30/2015] [Indexed: 12/31/2022]
|
9
|
Zhu K, Kakehi T, Matsumoto M, Iwata K, Ibi M, Ohshima Y, Zhang J, Liu J, Wen X, Taye A, Fan C, Katsuyama M, Sharma K, Yabe-Nishimura C. NADPH oxidase NOX1 is involved in activation of protein kinase C and premature senescence in early stage diabetic kidney. Free Radic Biol Med 2015; 83:21-30. [PMID: 25701431 DOI: 10.1016/j.freeradbiomed.2015.02.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 01/16/2015] [Accepted: 02/09/2015] [Indexed: 11/24/2022]
Abstract
Increased oxidative stress and activation of protein kinase C (PKC) under hyperglycemia have been implicated in the development of diabetic nephropathy. Because reactive oxygen species derived from nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, NOX1 accelerate the translocation of PKC isoforms, NOX1 is postulated to play a causative role in the development of diabetic nephropathy. Hyperglycemia was induced in wild-type and Nox1-deficient mice (KO) by two doses of streptozotocin injection. At 3 weeks after the induction of hyperglycemia, glomeruli and cortical tubules were isolated from kidneys. The mRNA level of Nox1 was significantly upregulated in the renal cortex at 3 weeks of hyperglycemia. Urinary albumin and expression of inflammatory or fibrotic mediators were similarly elevated in diabetic wild-type and KO; however, increases in glomerular volume and mesangial matrix area were attenuated in diabetic KO. Nox1 deficiency significantly reduced the levels of renal thiobarbituric acid-reacting substances and 8-hydroxydeoxyguanosine, membranous translocation of PKCα/β, activity of PKC, and phosphorylation of p38 mitogen-activated protein kinase in the diabetic kidney. Furthermore, increased staining of senescence-associated β-galactosidase in glomeruli and cortical tubules of diabetic mice was significantly suppressed in KO. Whereas the levels of cyclin-dependent kinase inhibitors, p16(INK4A) and p21(Cip1), were equivalent between the genotypes, increased levels of p27(Kip1) and γ-H2AX, a biomarker for DNA double-strand breaks, were significantly attenuated in isolated glomeruli and cortical tubules of diabetic KO. Taken together, NOX1 modulates the p38/p27(Kip1) signaling pathway by activating PKC and promotes premature senescence in early stage diabetic nephropathy.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tomoko Kakehi
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Misaki Matsumoto
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masakazu Ibi
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoichi Ohshima
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jia Zhang
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Junjie Liu
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Xiaopeng Wen
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ashraf Taye
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Chunyuan Fan
- Dujiangyan City Medical Center, Dujiangyan Chengdu, 611830 Sichuan, China
| | - Masato Katsuyama
- Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kumar Sharma
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Department of Medicine, University of California at San Diego and VA San Diego Healthcare System, La Jolla, CA 92093, USA
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| |
Collapse
|
10
|
Shah A, Xia L, Masson EAY, Gui C, Momen A, Shikatani EA, Husain M, Quaggin S, John R, Fantus IG. Thioredoxin-Interacting Protein Deficiency Protects against Diabetic Nephropathy. J Am Soc Nephrol 2015; 26:2963-77. [PMID: 25855771 DOI: 10.1681/asn.2014050528] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 01/22/2015] [Indexed: 12/24/2022] Open
Abstract
Expression of thioredoxin-interacting protein (TxNIP), an endogenous inhibitor of the thiol oxidoreductase thioredoxin, is augmented by high glucose (HG) and promotes oxidative stress. We previously reported that TxNIP-deficient mesangial cells showed protection from HG-induced reactive oxygen species, mitogen-activated protein kinase phosphorylation, and collagen expression. Here, we investigated the potential role of TxNIP in the pathogenesis of diabetic nephropathy (DN) in vivo. Wild-type (WT) control, TxNIP(-/-), and TxNIP(+/-) mice were rendered equally diabetic with low-dose streptozotocin. In contrast to effects in WT mice, diabetes did not increase albuminuria, proteinuria, serum cystatin C, or serum creatinine levels in TxNIP(-/-) mice. Whereas morphometric studies of kidneys revealed a thickened glomerular basement membrane and effaced podocytes in the diabetic WT mice, these changes were absent in the diabetic TxNIP(-/-) mice. Immunohistochemical analysis revealed significant increases in the levels of glomerular TGF-β1, collagen IV, and fibrosis only in WT diabetic mice. Additionally, only WT diabetic mice showed significant increases in oxidative stress (nitrotyrosine, urinary 8-hydroxy-2-deoxy-guanosine) and inflammation (IL-1β mRNA, F4/80 immunohistochemistry). Expression levels of Nox4-encoded mRNA and protein increased only in the diabetic WT animals. A significant loss of podocytes, assessed by Wilms' tumor 1 and nephrin staining and urinary nephrin concentration, was found in diabetic WT but not TxNIP(-/-) mice. Furthermore, in cultured human podocytes exposed to HG, TxNIP knockdown with siRNA abolished the increased mitochondrial O2 (-) generation and apoptosis. These data indicate that TxNIP has a critical role in the progression of DN and may be a promising therapeutic target.
Collapse
Affiliation(s)
- Anu Shah
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto General Research Institute, University Health Network, Department of Physiology, Banting and Best Diabetes Centre, and
| | - Ling Xia
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto General Research Institute, University Health Network, Banting and Best Diabetes Centre, and
| | - Elodie A Y Masson
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Banting and Best Diabetes Centre, and
| | - Chloe Gui
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Banting and Best Diabetes Centre, and
| | - Abdul Momen
- Toronto General Research Institute, University Health Network
| | - Eric A Shikatani
- Toronto General Research Institute, University Health Network, Department of Pathology and Laboratory Medicine, University of Toronto, Toronto, Ontario, Canada, and
| | - Mansoor Husain
- Toronto General Research Institute, University Health Network
| | - Susan Quaggin
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Banting and Best Diabetes Centre, and Feinberg Cardiovascular Research Institute, Division of Medicine-Nephrology, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Rohan John
- Toronto General Research Institute, University Health Network, Department of Pathology and Laboratory Medicine, University of Toronto, Toronto, Ontario, Canada, and
| | - I G Fantus
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto General Research Institute, University Health Network, Department of Physiology, Banting and Best Diabetes Centre, and
| |
Collapse
|
11
|
Jha JC, Gray SP, Barit D, Okabe J, El-Osta A, Namikoshi T, Thallas-Bonke V, Wingler K, Szyndralewiez C, Heitz F, Touyz RM, Cooper ME, Schmidt HHHW, Jandeleit-Dahm KA. Genetic targeting or pharmacologic inhibition of NADPH oxidase nox4 provides renoprotection in long-term diabetic nephropathy. J Am Soc Nephrol 2014; 25:1237-54. [PMID: 24511132 DOI: 10.1681/asn.2013070810] [Citation(s) in RCA: 297] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Diabetic nephropathy may occur, in part, as a result of intrarenal oxidative stress. NADPH oxidases comprise the only known dedicated reactive oxygen species (ROS)-forming enzyme family. In the rodent kidney, three isoforms of the catalytic subunit of NADPH oxidase are expressed (Nox1, Nox2, and Nox4). Here we show that Nox4 is the main source of renal ROS in a mouse model of diabetic nephropathy induced by streptozotocin administration in ApoE(-/-) mice. Deletion of Nox4, but not of Nox1, resulted in renal protection from glomerular injury as evidenced by attenuated albuminuria, preserved structure, reduced glomerular accumulation of extracellular matrix proteins, attenuated glomerular macrophage infiltration, and reduced renal expression of monocyte chemoattractant protein-1 and NF-κB in streptozotocin-induced diabetic ApoE(-/-) mice. Importantly, administration of the most specific Nox1/4 inhibitor, GKT137831, replicated these renoprotective effects of Nox4 deletion. In human podocytes, silencing of the Nox4 gene resulted in reduced production of ROS and downregulation of proinflammatory and profibrotic markers that are implicated in diabetic nephropathy. Collectively, these results identify Nox4 as a key source of ROS responsible for kidney injury in diabetes and provide proof of principle for an innovative small molecule approach to treat and/or prevent chronic kidney failure.
Collapse
Affiliation(s)
- Jay C Jha
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Stephen P Gray
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - David Barit
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Jun Okabe
- Human Epigenetics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Assam El-Osta
- Human Epigenetics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Tamehachi Namikoshi
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Japan
| | - Vicki Thallas-Bonke
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Kirstin Wingler
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Faculty of Medicine, Health & Life Science, Maastricht University, Maastricht, The Netherlands
| | | | | | - Rhian M Touyz
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mark E Cooper
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Harald H H W Schmidt
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Faculty of Medicine, Health & Life Science, Maastricht University, Maastricht, The Netherlands
| | - Karin A Jandeleit-Dahm
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
12
|
Malardé L, Rebillard A, Le Douairon-Lahaye S, Vincent S, Zguira MS, Lemoine-Morel S, Gratas-Delamarche A, Groussard C. Superoxide production pathways in aortas of diabetic rats: beneficial effects of insulin therapy and endurance training. Mol Cell Biochem 2013; 389:113-8. [PMID: 24374791 DOI: 10.1007/s11010-013-1932-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 12/18/2013] [Indexed: 12/31/2022]
Abstract
Superoxide (O 2 (·-) ) overproduction, by decreasing the nitric oxide ((·)NO) bioavailability, contributes to vascular complications in type 1 diabetes. In this disease, the vascular O 2 (·-) can be produced by the NADPH oxidase (NOX), nitric oxide synthase (NOS), and xanthine oxidase (XO). This study aimed to determine the contribution of each enzymatic pathway in hyperglycemia-induced O 2 (·-) overproduction, and the effects of an endurance training program and insulin therapy, associated or not, on the O 2 (·-) production (amount and related enzymes) in diabetic rats. Forty male Wistar rats were divided into diabetic (D), diabetic treated with insulin (D-Ins), diabetic trained (D-Tr), or diabetic insulin-treated and trained (D-Ins + Tr) groups. An additional healthy group was used as control. Insulin therapy (Glargine Lantus, Sanofi) and endurance training (treadmill run: 60 min/day, 25 m/min, 5 days/week) started 1 week after diabetes induction by streptozotocin (45 mg/kg), and lasted for 8 weeks. At the end of the protocol, the O 2 (·-) production in aorta rings was evaluated by histochemical analyses (DHE staining). Each production pathway was studied by inhibiting NOX (apocynin), NOS (L-Name), or XO (allopurinol) before DHE staining. Diabetic rats exhibited hyperglycemia-induced O 2 (·-) overproduction, resulting from NOX, NOS, and XO activation. Insulin therapy and endurance training, associated or not, decreased efficiently and similarly the O 2 (·-) overproduction. Insulin therapy reduced the hyperglycemia and decreased the three enzymatic pathways implicated in the O 2 (·-) production. Endurance training decreased directly the NOS and XO activity. While both therapeutic strategies activated different pathways, their association did not reduce the O 2 (·-) overproduction more significantly.
Collapse
Affiliation(s)
- L Malardé
- Laboratoire M2S, Université Rennes 2 - ENS Cachan, Avenue Robert Schuman, 35170, Bruz, France,
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Vázquez-Medina JP, Popovich I, Thorwald MA, Viscarra JA, Rodriguez R, Sonanez-Organis JG, Lam L, Peti-Peterdi J, Nakano D, Nishiyama A, Ortiz RM. Angiotensin receptor-mediated oxidative stress is associated with impaired cardiac redox signaling and mitochondrial function in insulin-resistant rats. Am J Physiol Heart Circ Physiol 2013; 305:H599-607. [PMID: 23771688 DOI: 10.1152/ajpheart.00101.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Activation of angiotensin receptor type 1 (AT1) contributes to NADPH oxidase (Nox)-derived oxidative stress during metabolic syndrome. However, the specific role of AT1 in modulating redox signaling, mitochondrial function, and oxidative stress in the heart remains more elusive. To test the hypothesis that AT1 activation increases oxidative stress while impairing redox signaling and mitochondrial function in the heart during diet-induced insulin resistance in obese animals, Otsuka Long Evans Tokushima Fatty (OLETF) rats (n = 8/group) were treated with the AT1 blocker (ARB) olmesartan for 6 wk. Cardiac Nox2 protein expression increased 40% in OLETF compared with age-matched, lean, strain-control Long Evans Tokushima Otsuka (LETO) rats, while mRNA and protein expression of the H₂O₂-producing Nox4 increased 40-100%. ARB treatment prevented the increase in Nox2 without altering Nox4. ARB treatment also normalized the increased levels of protein and lipid oxidation (nitrotyrosine, 4-hydroxynonenal) and increased the redox-sensitive transcription factor Nrf2 by 30% and the activity of antioxidant enzymes (SOD, catalase, GPx) by 50-70%. Citrate synthase (CS) and succinate dehydrogenase (SDH) activities decreased 60-70%, whereas cardiac succinate levels decreased 35% in OLETF compared with LETO, suggesting that mitochondrial function in the heart is impaired during obesity-induced insulin resistance. ARB treatment normalized CS and SDH activities, as well as succinate levels, while increasing AMPK and normalizing Akt, suggesting that AT1 activation also impairs cellular metabolism in the diabetic heart. These data suggest that the cardiovascular complications associated with metabolic syndrome may result from AT1 receptor-mediated Nox2 activation leading to impaired redox signaling, mitochondrial activity, and dysregulation of cellular metabolism in the heart.
Collapse
Affiliation(s)
- José Pablo Vázquez-Medina
- Department of Molecular and Cellular Biology, School of Natural Sciences, University of California Merced, Merced, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Choi HJ, Jang HJ, Chung TW, Jeong SI, Cha J, Choi JY, Han CW, Jang YS, Joo M, Jeong HS, Ha KT. Catalpol suppresses advanced glycation end-products-induced inflammatory responses through inhibition of reactive oxygen species in human monocytic THP-1 cells. Fitoterapia 2013; 86:19-28. [DOI: 10.1016/j.fitote.2013.01.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 11/25/2022]
|
15
|
Shah A, Xia L, Goldberg H, Lee KW, Quaggin SE, Fantus IG. Thioredoxin-interacting protein mediates high glucose-induced reactive oxygen species generation by mitochondria and the NADPH oxidase, Nox4, in mesangial cells. J Biol Chem 2013; 288:6835-48. [PMID: 23329835 DOI: 10.1074/jbc.m112.419101] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thioredoxin-interacting protein (TxNIP) is up-regulated by high glucose and is associated with oxidative stress. It has been implicated in hyperglycemia-induced β-cell dysfunction and apoptosis. As high glucose and oxidative stress mediate diabetic nephropathy (DN), the contribution of TxNIP was investigated in renal mesangial cell reactive oxygen species (ROS) generation and collagen synthesis. To determine the role of TxNIP, mouse mesangial cells (MC) cultured from wild-type C3H and TxNIP-deficient Hcb-19 mice were incubated in HG. Confocal microscopy was used to measure total and mitochondrial ROS production (DCF and MitoSOX) and collagen IV. Trx and NADPH oxidase activities were assayed and NADPH oxidase isoforms, Nox2 and Nox4, and antioxidant enzymes were determined by immunoblotting. C3H MC exposed to HG elicited a significant increase in cellular and mitochondrial ROS as well as Nox4 protein expression and NADPH oxidase activation, whereas Hcb-19 MC showed no response. Trx activity was attenuated by HG only in C3H MC. These defects in Hcb-19 MC were not due to increased antioxidant enzymes or scavenging of ROS, but associated with decreased ROS generation. Adenovirus-mediated overexpression of TxNIP in Hcb-19 MC and TxNIP knockdown with siRNA in C3H confirmed the specific role of TxNIP. Collagen IV accumulation in HG was markedly reduced in Hcb-19 cells. TxNIP is a critical component of the HG-ROS signaling pathway, required for the induction of mitochondrial and total cell ROS and the NADPH oxidase isoform, Nox4. TxNIP is a potential target to prevent DN.
Collapse
Affiliation(s)
- Anu Shah
- Department of Medicine and Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada
| | | | | | | | | | | |
Collapse
|
16
|
Dentelli P, Barale C, Togliatto G, Trombetta A, Olgasi C, Gili M, Riganti C, Toppino M, Brizzi MF. A diabetic milieu promotes OCT4 and NANOG production in human visceral-derived adipose stem cells. Diabetologia 2013; 56:173-84. [PMID: 23064289 DOI: 10.1007/s00125-012-2734-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 08/30/2012] [Indexed: 01/31/2023]
Abstract
AIMS/HYPOTHESIS Successful outcomes have been obtained by exploiting adipose-derived stem cells (ASCs) in regenerative medicine. NADPH oxidase (NOX)-generated reactive oxygen species (ROS) are known to control stem cell self-renewal. Several high glucose (HG)-mediated effects depend on NOX-generated ROS. In this study, we investigated whether, and how mechanistically, HG concentrations control ASC fate in patients with diabetes. METHODS ASCs from the visceral adipose tissue of non-diabetic (N-ASCs) and diabetic participants (D-ASCs), identified by surface markers, were counted and evaluated for ROS generation and stem cell properties. Their ability to release soluble factors was assessed by BioPlex analysis. To reproduce an in vitro diabetic glucose milieu, N-ASCs were cultured in HG (25 mmol/l) or normal glucose (NG) concentration (5 mmol/l), as control. ASC pluripotency was assessed by in vitro study. The p47(phox) NOX subunit, AKT and octamer-binding transcription factor 4 (OCT4; also known as POU5F1) were knocked down by small-interfering RNA technology. Stem-cell features were evaluated by sphere cluster formation. RESULTS The ASC number was higher in diabetic patients than in non-diabetic controls. Production of OCT4 and NANOG, stem-cell-specific transcription factors, was upregulated in D-ASCs compared with N-ASCs. Moreover, we found that ROS production and AKT activation drove D-ASC, but not N-ASC, secretion. When N-ASCs were cultured in vitro in the presence of HG, they also expressed OCT4/NANOG and formed spheres. By knock-down of the p47(phox) NOX subunit, AKT and OCT4 we demonstrated that NOX-generated ROS and their downstream signals are crucial for HG-mediated ASC de-differentiation and proinflammatory cytokine production. CONCLUSIONS/INTERPRETATION We herein provide a rationale for exploiting D-ASCs in regenerative medicine and/or exploiting HG preconditioning to increase ASCs ex vivo.
Collapse
Affiliation(s)
- P Dentelli
- Department of Medical Sciences, University of Torino, Corso Dogliotti 14, 10126 Torino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fazeli G, Stopper H, Schinzel R, Ni CW, Jo H, Schupp N. Angiotensin II induces DNA damage via AT1 receptor and NADPH oxidase isoform Nox4. Mutagenesis 2012; 27:673-81. [PMID: 22844079 PMCID: PMC6281047 DOI: 10.1093/mutage/ges033] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epidemiological studies revealed increased renal cancer incidences and higher cancer mortalities in hypertensive individuals. Activation of the renin-angiotensin-aldosterone system leads to the formation of reactive oxygen species (ROS). In vitro, in renal cells, and ex vivo, in the isolated perfused mouse kidney, we could show DNA-damaging potential of angiotensin II (Ang II). Here, the pathway involved in the genotoxicity of Ang II was investigated. In kidney cell lines with properties of proximal tubulus cells, an activation of NADPH oxidase and the production of ROS, resulting in the formation of DNA strand breaks and micronuclei induction, was observed. This DNA damage was mediated by the Ang II type 1 receptor (AT1R), together with the G protein G ( α-q/11 ) . Subsequently, phospholipase C (PLC) was activated and intracellular calcium increased. Both calcium stores of the endoplasmic reticulum and extracellular calcium were involved in the genotoxicity of Ang II. Downstream, a role for protein kinase C (PKC) could be detected, because its inhibition hindered Ang II from damaging the cells. Although PKC was activated, no involvement of its known target, the NADPH oxidase isoform containing the Nox2 subunit, could be found, as tested by small-interfering RNA down-regulation. Responsible for the DNA-damaging activity of Ang II was the NADPH oxidase isoform containing the Nox4 subunit. In summary, in kidney cells the DNA-damaging activity of Ang II depends on an AT1R-mediated activation of NADPH oxidase via PLC, PKC and calcium signalling, with the NADPH subunit Nox4 playing a crucial role.
Collapse
Affiliation(s)
- Gholamreza Fazeli
- Institute of Pharmacology and Toxicology, Versbacher Str. 9, University of Würzburg, 97078 Würzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, Versbacher Str. 9, University of Würzburg, 97078 Würzburg, Germany
| | | | - Chih-Wen Ni
- Coulter Department of Biomedical EngineeringGeorgia Tech and Emory University, Atlanta, Georgia 30322, USA
| | - Hanjoong Jo
- Coulter Department of Biomedical EngineeringGeorgia Tech and Emory University, Atlanta, Georgia 30322, USA
| | - Nicole Schupp
- Institute of Pharmacology and Toxicology, Versbacher Str. 9, University of Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
18
|
Is oxidative stress, a link between nephrolithiasis and obesity, hypertension, diabetes, chronic kidney disease, metabolic syndrome? ACTA ACUST UNITED AC 2012; 40:95-112. [PMID: 22213019 DOI: 10.1007/s00240-011-0448-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 12/10/2011] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have provided the evidence for association between nephrolithiasis and a number of cardiovascular diseases including hypertension, diabetes, chronic kidney disease, metabolic syndrome. Many of the co-morbidities may not only lead to stone disease but also be triggered by it. Nephrolithiasis is a risk factor for development of hypertension and have higher prevalence of diabetes mellitus and some hypertensive and diabetic patients are at greater risk for stone formation. An analysis of the association between stone disease and other simultaneously appearing disorders, as well as factors involved in their pathogenesis, may provide an insight into stone formation and improved therapies for stone recurrence and prevention. It is our hypothesis that association between stone formation and development of co-morbidities is a result of certain common pathological features. Review of the recent literature indicates that production of reactive oxygen species (ROS) and development of oxidative stress (OS) may be such a common pathway. OS is a common feature of all cardiovascular diseases (CVD) including hypertension, diabetes mellitus, atherosclerosis and myocardial infarct. There is increasing evidence that ROS are also produced during idiopathic calcium oxalate (CaOx) nephrolithiasis. Both tissue culture and animal model studies demonstrate that ROS are produced during interaction between CaOx/calcium phosphate (CaP) crystals and renal epithelial cells. Clinical studies have also provided evidence for the development of oxidative stress in the kidneys of stone forming patients. Renal disorders which lead to OS appear to be a continuum. Stress produced by one disorder may trigger the other under the right circumstances.
Collapse
|
19
|
Targeting endothelial dysfunction in vascular complications associated with diabetes. Int J Vasc Med 2011; 2012:750126. [PMID: 22013533 PMCID: PMC3195347 DOI: 10.1155/2012/750126] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/04/2011] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular complications associated with diabetes remain a significant health issue in westernized societies. Overwhelming evidence from clinical and laboratory investigations have demonstrated that these cardiovascular complications are initiated by a dysfunctional vascular endothelium. Indeed, endothelial dysfunction is one of the key events that occur during diabetes, leading to the acceleration of cardiovascular mortality and morbidity. In a diabetic milieu, endothelial dysfunction occurs as a result of attenuated production of endothelial derived nitric oxide (EDNO) and augmented levels of reactive oxygen species (ROS). Thus, in this review, we discuss novel therapeutic targets that either upregulate EDNO production or increase antioxidant enzyme capacity in an effort to limit oxidative stress and restore endothelial function. In particular, endogenous signaling molecules that positively modulate EDNO synthesis and mimetics of endogenous antioxidant enzymes will be highlighted. Consequently, manipulation of these unique targets, either alone or in combination, may represent a novel strategy to confer vascular protection, with the ultimate goal of improved outcomes for diabetes-associated vascular complications.
Collapse
|
20
|
Simvastatin protects osteoblast against H2O2-induced oxidative damage via inhibiting the upregulation of Nox4. Mol Cell Biochem 2011; 360:71-7. [DOI: 10.1007/s11010-011-1045-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 08/27/2011] [Indexed: 11/26/2022]
|
21
|
Piwkowska A, Rogacka D, Audzeyenka I, Jankowski M, Angielski S. High glucose concentration affects the oxidant-antioxidant balance in cultured mouse podocytes. J Cell Biochem 2011; 112:1661-72. [PMID: 21503956 DOI: 10.1002/jcb.23088] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hyperglycemia is well-recognized and has long-term complications in diabetes mellitus and diabetic nephropathy. In podocytes, the main component of the glomerular barrier, overproduction of reactive oxygen species (ROS) in the presence of high glucose induces dysfunction and increases excretion of albumin in urine. This suggests an impaired antioxidant defense system has a role in the pathogenesis of diabetic nephropathy. We studied expression of NAD(P)H oxidase subunits by Western blotting and immunofluorescence and the activities of the oxidant enzyme, NAD(P)H, and antioxidant enzymes, superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT), in mouse podocytes cultured in a high glucose concentration (30 mM). We found long-term (3 and 5 days) exposure of mouse podocytes to high glucose concentrations caused oxidative stress, as evidenced by increased expression of Nox4 and activities of NAD(P)H oxidase (Δ 182%) and SOD (Δ 39%) and decreased activities of GPx (Δ -40%) and CAT (Δ -35%). These biochemical changes were accompanied by a rise in intracellular ROS production and accumulation of hydrogen peroxide in extracellular space. The role of Nox4 in ROS generation was confirmed with Nox4 siRNA. In conclusion, high glucose concentration affects the oxidant-antioxidant balance in mouse podocytes, resulting in enhanced generation of superoxide anions and its attenuated metabolism. These observations suggest free radicals may play an important role in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Agnieszka Piwkowska
- Mossakowski Medical Research Center Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland.
| | | | | | | | | |
Collapse
|
22
|
Abstract
The number of people with diabetic kidney disease continues to increase worldwide despite current treatments. Of the pathophysiologic mechanisms that have been identified in the development and progression of diabetic nephropathy, oxidative stress (more accurately described as increased levels of reactive oxygen species; ROS) is of major importance. The increase in ROS is due to both increased production and to decreased and/or inadequate antioxidant function. To date, human clinical trials with antioxidants have not been shown to be effective. This is likely due, at least in part, to the lack of specificity of current agents. Recent research has determined both major sources of high glucose-induced cellular ROS production as well as high glucose-induced changes in antioxidant function. Treatments targeted at one or more of the specific diabetes-induced alterations in the regulation of ROS levels will likely lead to effective treatments that prevent the development and progression of diabetic kidney disease.
Collapse
Affiliation(s)
- Robert C Stanton
- Harvard Medical School, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA.
| |
Collapse
|
23
|
Tabet F, Lambert G, Cuesta Torres LF, Hou L, Sotirchos I, Touyz RM, Jenkins AJ, Barter PJ, Rye KA. Lipid-Free Apolipoprotein A-I and Discoidal Reconstituted High-Density Lipoproteins Differentially Inhibit Glucose-Induced Oxidative Stress in Human Macrophages. Arterioscler Thromb Vasc Biol 2011; 31:1192-200. [DOI: 10.1161/atvbaha.110.222000] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective—
The goal of this study was to investigate the mechanisms by which apolipoprotein (apo) A-I, in the lipid-free form or as a constituent of discoidal reconstituted high-density lipoproteins ([A-I]rHDL), inhibits high-glucose–induced redox signaling in human monocyte-derived macrophages (HMDM).
Methods and Results—
HMDM were incubated under normal (5.8 mmol/L) or high-glucose (25 mmol/L) conditions with native high-density lipoproteins (HDL) lipid-free apoA-I from normal subjects and from subjects with type 2 diabetes (T2D) or (A-I)rHDL. Superoxide (O
2
−
) production was measured using dihydroethidium fluorescence. NADPH oxidase activity was assessed using lucigenin-derived chemiluminescence and a cyotochrome
c
assay. p47phox translocation to the plasma membrane, Nox2, superoxide dismutase 1 (SOD1), and SOD2 mRNA and protein levels were determined by real-time polymerase chain reaction and Western blotting. Native HDL induced a time-dependent inhibition of O
2
−
generation in HMDM incubated with 25 mmol/L glucose. Lipid-free apoA-I and (A-I)rHDL increased SOD1 and SOD2 levels and attenuated 25 mmol/L glucose-mediated increases in cellular O
2
−
, NADPH oxidase activity, p47 translocation, and Nox2 expression. Lipid-free apoA-I mediated its effects on Nox2, SOD1, and SOD2 via ABCA1. (A-I)rHDL-mediated effects were via ABCG1 and scavenger receptor BI. Lipid-free apoA-I from subjects with T2D inhibited reactive oxygen species generation less efficiently than normal apoA-I.
Conclusion—
Native HDL, lipid-free apoA-I and (A-I)rHDL inhibit high-glucose–induced redox signaling in HMDM. The antioxidant properties of apoA-I are attenuated in T2D.
Collapse
Affiliation(s)
- Fatiha Tabet
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Gilles Lambert
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Luisa F. Cuesta Torres
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Liming Hou
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Irene Sotirchos
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Rhian M. Touyz
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Alicia J. Jenkins
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Philip J. Barter
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| | - Kerry-Anne Rye
- From the Lipid Research Group, Heart Research Institute, Sydney, New South Wales, Australia (F.T., G.L., L.F.C.T., L.H., I.S., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (P.J.B., K.-A.R.); Faculté de Médecine, Université de Nantes, Institut National de la Santé et de la Recherche Médicale U957, Nantes, France (G.L.); Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (R.M.T.); Department of Medicine, University of
| |
Collapse
|
24
|
Randazzo J, Zhang P, Makita J, Blessing K, Kador PF. Orally active multi-functional antioxidants delay cataract formation in streptozotocin (type 1) diabetic and gamma-irradiated rats. PLoS One 2011; 6:e18980. [PMID: 21541328 PMCID: PMC3082543 DOI: 10.1371/journal.pone.0018980] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 03/21/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Age-related cataract is a worldwide health care problem whose progression has been linked to oxidative stress and the accumulation of redox-active metals. Since there is no specific animal model for human age-related cataract, multiple animal models must be used to evaluate potential therapies that may delay and/or prevent cataract formation. METHODS/PRINCIPAL FINDINGS Proof of concept studies were conducted to evaluate 4-(5-hydroxypyrimidin-2-yl)-N,N-dimethyl-3,5-dioxopiperazine-1-sulfonamide (compound 4) and 4-(5-hydroxy-4,6-dimethoxypyrimidin-2-yl)-N,N-dimethyl-3,5-dioxopiperazine-1-sulfonamide (compound 8), multi-functional antioxidants that can independently chelate redox metals and quench free radicals, on their ability to delay the progression of diabetic "sugar" cataracts and gamma radiation-induced cataracts. Prior to 15 Gy of whole head irradiation, select groups of Long Evans rats received either diet containing compound 4 or 8, or a single i.p. injection of panthethine, a radioprotective agent. Compared to untreated, irradiated rats, treatment with pantethine, 4 and 8 delayed initial lens changes by 4, 47, and 38 days, respectively, and the average formation of posterior subcapsular opacities by 23, 53 and 58 days, respectively. In the second study, select groups of diabetic Sprague Dawley rats were administered chow containing compounds 4, 8 or the aldose reductase inhibitor AL1576. As anticipated, treatment with AL1576 prevented cataract by inhibiting sorbitol formation in the lens. However, compared to untreated rats, compounds 4 and 8 delayed vacuole formation by 20 days and 12 days, respectively, and cortical cataract formation by 8 and 3 days, respectively, without reducing lenticular sorbitol. Using in vitro lens culture in 30 mM xylose to model diabetic "sugar" cataract formation, western blots confirmed that multi-functional antioxidants reduced endoplasmic reticulum stress. CONCLUSIONS/SIGNIFICANCE Multi-functional antioxidants delayed cataract formation in two diverse rat models. These studies provide a proof of concept that a general cataract treatment focused on reducing oxidative stress instead of a specific mechanism of cataractogenesis can be developed.
Collapse
Affiliation(s)
- James Randazzo
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Peng Zhang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jun Makita
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Karen Blessing
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Peter F. Kador
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
25
|
Park JB. Identification and quantification of a major anti-oxidant and anti-inflammatory phenolic compound found in basil, lemon thyme, mint, oregano, rosemary, sage, and thyme. Int J Food Sci Nutr 2011; 62:577-84. [DOI: 10.3109/09637486.2011.562882] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Park J, Kwon MK, Huh JY, Choi WJ, Jeong LS, Nagai R, Kim WY, Kim J, Lee GT, Lee HB, Ha H. Renoprotective antioxidant effect of alagebrium in experimental diabetes. Nephrol Dial Transplant 2011; 26:3474-84. [DOI: 10.1093/ndt/gfr152] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
27
|
Piwkowska A, Rogacka D, Jankowski M, Dominiczak MH, Stepiński JK, Angielski S. Metformin induces suppression of NAD(P)H oxidase activity in podocytes. Biochem Biophys Res Commun 2010; 393:268-73. [PMID: 20123087 DOI: 10.1016/j.bbrc.2010.01.119] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 01/28/2010] [Indexed: 11/30/2022]
Abstract
Hyperglycemia increases the production of reactive oxygen species (ROS). NAD(P)H oxidase, producing superoxide anion, is the main source of ROS in diabetic podocytes and their production contributes to the development of diabetic nephropathy. We have investigated the effect of an antidiabetic drug, metformin on the production of superoxide anion in cultured podocytes and attempted to elucidate underlying mechanisms. The experiments were performed in normal (NG, 5.6mM) and high (HG, 30mM) glucose concentration. Overall ROS production was measured by fluorescence of a DCF probe. Activity of NAD(P)H oxidase was measured by chemiluminescence method. The AMP-dependent kinase (AMPK) activity was determined by immunobloting, measuring the ratio of phosphorylated AMPK to total AMPK. Glucose accumulation was measured using 2-deoxy-[1,2-(3)H]-glucose. ROS production increased by about 27% (187+/-8 vs. 238+/-9 arbitrary units AU, P<0.01) in HG. Metformin (2mM, 2h) markedly reduced ROS production by 45% in NG and 60% in HG. Metformin decreased NAD(P)H oxidase activity in NG (36%) and HG (86%). AMPK activity was increased by metformin in NG and HG (from 0.58+/-0.07 to. 0.99+/-0.06, and from 0.53+/-0.03 to 0.64+/-0.03; P<0.05). The effects of metformin on the activities of NAD(P)H oxidase and AMPK were abolished in the presence of AMPK inhibitor, compound C. We have shown that metformin decreases production of ROS through reduction of NAD(P)H oxidase activity. We also have demonstrated relationship between activity of NAD(P)H oxidase and AMPK.
Collapse
Affiliation(s)
- Agnieszka Piwkowska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Gdańsk, Poland.
| | | | | | | | | | | |
Collapse
|
28
|
Cui W, Matsuno K, Iwata K, Ibi M, Katsuyama M, Kakehi T, Sasaki M, Ikami K, Zhu K, Yabe-Nishimura C. NADPH Oxidase Isoforms and Anti-hypertensive Effects of Atorvastatin Demonstrated in Two Animal Models. J Pharmacol Sci 2009; 111:260-8. [DOI: 10.1254/jphs.09148fp] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|