51
|
Khoury T, Tzukert K, Abel R, Abu Rmeileh A, Levi R, Ilan Y. The gut-kidney axis in chronic renal failure: A new potential target for therapy. Hemodial Int 2017; 21:323-334. [PMID: 27634464 DOI: 10.1111/hdi.12486] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 08/06/2016] [Indexed: 12/26/2022]
Abstract
Evidence is accumulating to consider the gut microbiome as a central player in the gut-kidney axis. Microbiome products, such as advanced glycation end products, phenols, and indoles, are absorbed into the circulation but are cleared by normal-functioning kidneys. These products then become toxic and contribute to the uremic load and to the progression of chronic kidney failure. In this review, we discuss the gut-kidney interaction under the state of chronic kidney failure as well as the potential mechanisms by which a change in the gut flora (termed gut dysbiosis) in chronic kidney disease (CKD) exacerbates uremia and leads to further progression of CKD and inflammation. Finally, the potential therapeutic interventions to target the gut microbiome in CKD are discussed.
Collapse
Affiliation(s)
- Tawfik Khoury
- Department of Medicine, Gastroenterology and Liver Units, Jerusalem, Israel
| | - Keren Tzukert
- Department of Nephrology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Roy Abel
- Department of Nephrology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Ayman Abu Rmeileh
- Department of Medicine, Gastroenterology and Liver Units, Jerusalem, Israel
| | - Ronen Levi
- Department of Nephrology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Department of Medicine, Gastroenterology and Liver Units, Jerusalem, Israel
| |
Collapse
|
52
|
Sex and gender differences in chronic kidney disease: progression to end-stage renal disease and haemodialysis. Clin Sci (Lond) 2017; 130:1147-63. [PMID: 27252402 DOI: 10.1042/cs20160047] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/29/2016] [Indexed: 01/04/2023]
Abstract
Sex and gender differences are of fundamental importance in most diseases, including chronic kidney disease (CKD). Men and women with CKD differ with regard to the underlying pathophysiology of the disease and its complications, present different symptoms and signs, respond differently to therapy and tolerate/cope with the disease differently. Yet an approach using gender in the prevention and treatment of CKD, implementation of clinical practice guidelines and in research has been largely neglected. The present review highlights some sex- and gender-specific evidence in the field of CKD, starting with a critical appraisal of the lack of inclusion of women in randomized clinical trials in nephrology, and thereafter revisits sex/gender differences in kidney pathophysiology, kidney disease progression, outcomes and management of haemodialysis care. In each case we critically consider whether apparent discrepancies are likely to be explained by biological or psycho-socioeconomic factors. In some cases (a few), these findings have resulted in the discovery of disease pathways and/or therapeutic opportunities for improvement. In most cases, they have been reported as merely anecdotal findings. The aim of the present review is to expose some of the stimulating hypotheses arising from these observations as a preamble for stricter approaches using gender for the prevention and treatment of CKD and its complications.
Collapse
|
53
|
Guo J, Lu L, Hua Y, Huang K, Wang I, Huang L, Fu Q, Chen A, Chan P, Fan H, Liu ZM, Wang BH. Vasculopathy in the setting of cardiorenal syndrome: roles of protein-bound uremic toxins. Am J Physiol Heart Circ Physiol 2017; 313:H1-H13. [PMID: 28411233 DOI: 10.1152/ajpheart.00787.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) often leads to and accelerates the progression of cardiovascular disease (CVD), while CVD also causes kidney dysfunction. This bidirectional interaction leads to the development of a complex syndrome known as cardiorenal syndrome (CRS). CRS not only involves both the heart and the kidney but also the vascular system through a vast array of contributing factors. In addition to hemodynamic, neurohormonal, mechanical, and biochemical factors, nondialyzable protein-bound uremic toxins (PBUTs) are also key contributing factors that have been demonstrated through in vitro, in vivo, and clinical observations. PBUTs are ineffectively removed by hemodialysis because their complexes with albumins are larger than the pores of the dialysis membranes. PBUTs such as indoxyl sulfate and p-cresyl sulfate are key determinate and predictive factors for the progression of CVD in CKD patients. In CRS, both vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) exhibit significant dysfunction that is associated with the progression of CVD. PBUTs influence proliferation, calcification, senescence, migration, inflammation, and oxidative stress in VSMCs and ECs through various mechanisms. These pathological changes lead to arterial remodeling, stiffness, and atherosclerosis and thus reduce heart perfusion and impair left ventricular function, aggravating CRS. There is limited literature about the effect of PBUT on the vascular system and their contribution to CRS. This review summarizes current knowledge on how PBUTs influence vasculature, clarifies the relationship between uremic toxin-related vascular disease and CRS, and highlights the potential therapeutic strategies of uremic vasculopathy in the setting of CRS.
Collapse
Affiliation(s)
- Jingbin Guo
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.,Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Diseases, Guangzhou, China
| | - Lu Lu
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yue Hua
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Kevin Huang
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ian Wang
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia;
| | - Li Huang
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Qiang Fu
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Diseases, Guangzhou, China
| | - Aihua Chen
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Diseases, Guangzhou, China
| | - Paul Chan
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai, China; and.,Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Huimin Fan
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai, China; and
| | - Zhong-Min Liu
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai, China; and
| | - Bing Hui Wang
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
54
|
Wang LL, Huang YH, Yan CY, Wei XD, Hou JQ, Pu JX, Lv JX. N-acetylcysteine Ameliorates Prostatitis via miR-141 Regulating Keap1/Nrf2 Signaling. Inflammation 2017; 39:938-47. [PMID: 26941030 DOI: 10.1007/s10753-016-0327-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic prostatitis was the most common type of prostatitis and oxidative stress was reported to be highly elevated in prostatitis patients. In this study, we determined the effect of N-acetylcysteine (NAC) on prostatitis and the molecular mechanism involved in it. Male Sprague-Dawley rats were divided into three groups: control group (group A, n = 20), carrageenan-induced chronic nonbacterial prostatitis (CNP) model group (group B, n = 20), and carrageenan-induced CNP model group with NAC injection (group C, n = 20). Eye score, locomotion score, inflammatory cell count, cyclooxygenase 2 (COX2) expression, and Evans blue were compared in these three groups. The expression of miR-141 was determined by quantitative real-time PCR (qRT-PCR). Moreover, protein expressions of Kelch-like ECH-associated protein-1 (Keap1) and nuclear factor erythroid-2 related factor 2 (Nrf2) and its target genes were examined by Western blot. Luciferase reporter assay was performed in RWPE-1 cells transfected miR-141 mimic or inhibitor and the plasmid carrying 3'-UTR of Keap1. The value of eye score, locomotion score, inflammatory cell count, and Evans blue were significantly decreased in group C, as well as the expression of COX2, when comparing to that of group B. These results indicated that NAC relieved the carrageenan-induced CNP. Further, we found that NAC increased the expression of miR-141 and activated the Keap1/Nrf2 signaling. Luciferase reporter assay revealed that miR-141 mimic could suppress the activity of Keap1 and stimulate the downstream target genes of Nrf2. In addition, miR-141 inhibitor could reduce the effect of NAC on prostatitis. NAC ameliorates the carrageenan-induced prostatitis and prostate inflammation pain through miR-141 regulating Keap1/Nrf2 signaling.
Collapse
Affiliation(s)
- Liang-Liang Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu-Hua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun-Yin Yan
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xue-Dong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Quan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin-Xian Pu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin-Xing Lv
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, No.188, Shizi St, Canglang District, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
55
|
A polymorphism in a major antioxidant gene (Kelch-like ECH-associated protein 1) predicts incident cardiovascular events in chronic kidney disease patients: an exploratory study. J Hypertens 2016; 34:928-34. [PMID: 26974313 DOI: 10.1097/hjh.0000000000000878] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Oxidative stress is considered a major pathway conducive to cardiovascular disease in chronic kidney disease (CKD) patients. However, observational studies and clinical trials testing this relationship are controversial. The Nuclear factor-erythroid-2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) system is a major system regulating antioxidant mechanisms in living organisms. Owing to the fact that genes are transmitted randomly (Mendelian randomization), genetic variants may provide unconfounded assessment of putative causal risk factors. METHODS We have therefore explored the association of eight polymorphisms in the Nrf2 gene and three polymorphisms in the Keap1 gene (capturing over 80% of the genetic variance in the same genes) with cardiovascular events in a multicenter cohort study of 758 CKD patients. RESULTS During the follow-up period, 117 patients had fatal and nonfatal cardiovascular events and 42 died. The hazard rate of fatal and nonfatal cardiovascular outcomes was about twice higher in patients with the AA or the CA genotype (dominant model) in the rs110857735 polymorphism of the Keap1 gene (hazard rate: 1.85, 95% CI: 1.20-2.84, P = 0.005) than in those with the CC genotype. Further analyses adjusting for Framingham risk factors and CKD-specific risk factors and a bootstrapping validation analysis did not modify the strength of this association. No association was registered between other Keap1 and Nrf2 polymorphisms and cardiovascular disease in the same cohort. CONCLUSION In this exploratory study a gene-variant in Keap1, a major gene regulating the antioxidant response, predicts incident cardiovascular events in CKD patients. This finding is in keeping with the hypothesis implicating oxidative stress in cardiovascular disease in this population.
Collapse
|
56
|
Toya K, Babazono T, Murata H, Hanai K, Uchigata Y. Association of serum bilirubin levels with mortality in patients with diabetes initiating chronic hemodialysis: a competing risks analysis of a single-center cohort. RENAL REPLACEMENT THERAPY 2016. [DOI: 10.1186/s41100-016-0065-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
57
|
Park B, Mavanur A, Drezner AD, Gallagher J, Menzoian JO. Clinical Impact of Chronic Renal Insufficiency on Endovascular Aneurysm Repair. Vasc Endovascular Surg 2016; 40:437-45. [PMID: 17202089 DOI: 10.1177/1538574406294071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endovascular aneurysm repair of abdominal aortic aneurysms has become a viable alternative to open repair. A significant proportion of this patient population has chronic renal insufficiency. The surgical outcomes associated with endovascular repair in 342 patients, with and without chronic renal insufficiency, are reported. Perioperative mortality, length of admission, length of intensive care unit admission, and rates of acute renal failure, congestive heart failure, myocardial infarction, conversion to open surgery, progression to hemodialysis, and incidence of endoleaks were retrospectively reviewed and analyzed. Endovascular repair demonstrated higher rates of acute renal failure, longer length of stay, and longer intensive care unit admissions in patients with chronic renal insufficiency. Patients with severe renal dysfunction demonstrated markedly elevated mortality and morbidity. These results indicate that chronic renal insufficiency is not an absolute contraindication to endovascular repair in patients with moderate renal dysfunction, but patients with severe renal dysfunction perform poorly after aortic reconstruction.
Collapse
Affiliation(s)
- Brian Park
- Division of Vascular Surgery, Hartford Hospital, Hartford, Connecticut 06102-5037, USA
| | | | | | | | | |
Collapse
|
58
|
Koppe L, Nyam E, Vivot K, Manning Fox JE, Dai XQ, Nguyen BN, Trudel D, Attané C, Moullé VS, MacDonald PE, Ghislain J, Poitout V. Urea impairs β cell glycolysis and insulin secretion in chronic kidney disease. J Clin Invest 2016; 126:3598-612. [PMID: 27525435 DOI: 10.1172/jci86181] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/24/2016] [Indexed: 12/25/2022] Open
Abstract
Disorders of glucose homeostasis are common in chronic kidney disease (CKD) and are associated with increased mortality, but the mechanisms of impaired insulin secretion in this disease remain unclear. Here, we tested the hypothesis that defective insulin secretion in CKD is caused by a direct effect of urea on pancreatic β cells. In a murine model in which CKD is induced by 5/6 nephrectomy (CKD mice), we observed defects in glucose-stimulated insulin secretion in vivo and in isolated islets. Similarly, insulin secretion was impaired in normal mouse and human islets that were cultured with disease-relevant concentrations of urea and in islets from normal mice treated orally with urea for 3 weeks. In CKD mouse islets as well as urea-exposed normal islets, we observed an increase in oxidative stress and protein O-GlcNAcylation. Protein O-GlcNAcylation was also observed in pancreatic sections from CKD patients. Impairment of insulin secretion in both CKD mouse and urea-exposed islets was associated with reduced glucose utilization and activity of phosphofructokinase 1 (PFK-1), which could be reversed by inhibiting O-GlcNAcylation. Inhibition of O-GlcNAcylation also restored insulin secretion in both mouse models. These results suggest that insulin secretory defects associated with CKD arise from elevated circulating levels of urea that increase islet protein O-GlcNAcylation and impair glycolysis.
Collapse
|
59
|
Nakayama M, Kabayama S, Ito S. The hydrogen molecule as antioxidant therapy: clinical application in hemodialysis and perspectives. RENAL REPLACEMENT THERAPY 2016. [DOI: 10.1186/s41100-016-0036-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
60
|
Nigwekar SU, Kang A, Zoungas S, Cass A, Gallagher MP, Kulshrestha S, Navaneethan SD, Perkovic V, Strippoli GFM, Jardine MJ, Cochrane Kidney and Transplant Group. Interventions for lowering plasma homocysteine levels in dialysis patients. Cochrane Database Syst Rev 2016; 2016:CD004683. [PMID: 27243372 PMCID: PMC8520736 DOI: 10.1002/14651858.cd004683.pub4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND People with end-stage kidney disease (ESKD) have high rates of cardiovascular events. Randomised controlled trials (RCTs) of homocysteine-lowering therapies have not shown reductions in cardiovascular event rates in the general population. However, people with kidney disease have higher levels of homocysteine and may have different mechanisms of cardiovascular disease. We performed a systematic review of the effect of homocysteine-lowering therapies in people with ESKD. OBJECTIVES To evaluate the benefits and harms of established homocysteine lowering therapy (folic acid, vitamin B6, vitamin B12) on all-cause mortality and cardiovascular event rates in patients with ESKD. SEARCH METHODS We searched Cochrane Kidney and Transplant's Specialised Register to 25 January 2016 through contact with the Information Specialist using search terms relevant to this review. SELECTION CRITERIA Studies conducted in people with ESKD that reported at least 100 patient-years of follow-up and assessed the effect of therapies that are known to have homocysteine-lowering properties were included. DATA COLLECTION AND ANALYSIS Two authors independently extracted data using a standardised form. The primary outcome was cardiovascular mortality. Secondary outcomes included all-cause mortality, incident cardiovascular disease (fatal and nonfatal myocardial infarction and coronary revascularisation), cerebrovascular disease (stroke and cerebrovascular revascularisation), peripheral vascular disease (lower limb amputation), venous thromboembolic disease (deep vein thrombosis and pulmonary embolism), thrombosis of dialysis access, and adverse events. The effects of homocysteine-lowering therapies on outcomes were assessed with meta-analyses using random-effects models. Prespecified subgroup and sensitivity analyses were conducted. MAIN RESULTS We included six studies that reported data on 2452 participants with ESKD. Interventions investigated were folic acid with or without other vitamins (vitamin B6, vitamin B12). Participants' mean age was 48 to 65 years, and proportions of male participants ranged from 50% to 98%.Homocysteine-lowering therapy probably leads to little or no effect on cardiovascular mortality (4 studies, 1186 participants: RR 0.93, 95% CI 0.70 to 1.22). There was no evidence of heterogeneity among the included studies (I² = 0%). Homocysteine-lowering therapy had little or no effect on all-cause mortality or any other of this review's secondary outcomes. All prespecified subgroup and sensitivity analyses demonstrated little or no difference. Reported adverse events were mild and there was no increase in the incidence of adverse events from homocysteine-lowering therapies (3 studies, 1248 participants: RR 1.12, 95% CI 0.51 to 2.47; I(2) = 0%). Overall, studies were assessed as being at low risk of bias and there was no evidence of publication bias. AUTHORS' CONCLUSIONS Homocysteine-lowering therapies were not found to reduce mortality (cardiovascular and all-cause) or cardiovascular events among people with ESKD.
Collapse
Affiliation(s)
- Sagar U Nigwekar
- Harvard Medical SchoolDivision of Nephrology, Massachusetts General Hospital, Scholars in Clinical Sciences ProgramBostonMAUSA
| | - Amy Kang
- The University of SydneySydney Medical SchoolSydneyNSWAustralia
- The George Institute for Global Health, The University of SydneyRenal and Metabolic DivisionCamperdownNSWAustralia
| | - Sophia Zoungas
- Monash UniversityDiabetes and Vascular Research Program, Monash Centre for Health Research and Implementation, School of Public Health and Preventive MedicineClaytonVICAustralia
| | - Alan Cass
- The George Institute for Global Health, The University of SydneyRenal and Metabolic DivisionCamperdownNSWAustralia
- Menzies School of Health ResearchPO Box 41096CasuarinaNTAustralia0811
| | - Martin P Gallagher
- The George Institute for Global Health, The University of SydneyRenal and Metabolic DivisionCamperdownNSWAustralia
| | - Satyarth Kulshrestha
- University of Iowa Carver College of MedicineDepartment of Nephrology200 Hawkins Drive‐T307GHIowa CityIAUSA52242
| | | | - Vlado Perkovic
- The George Institute for Global Health, The University of SydneyRenal and Metabolic DivisionCamperdownNSWAustralia
| | - Giovanni FM Strippoli
- The Children's Hospital at WestmeadCochrane Kidney and Transplant, Centre for Kidney ResearchWestmeadNSWAustralia2145
- University of BariDepartment of Emergency and Organ TransplantationBariItaly
- DiaverumMedical Scientific OfficeLundSweden
- Diaverum AcademyBariItaly
- The University of SydneySydney School of Public HealthSydneyAustralia
| | - Meg J Jardine
- The George Institute for Global Health, The University of SydneyRenal and Metabolic DivisionCamperdownNSWAustralia
- Concord Repatriation General HospitalDepartment of Renal MedicineHospital RoadConcordNSWAustralia2139
| | | |
Collapse
|
61
|
Dey S, Sidor A, O'Rourke B. Compartment-specific Control of Reactive Oxygen Species Scavenging by Antioxidant Pathway Enzymes. J Biol Chem 2016; 291:11185-97. [PMID: 27048652 PMCID: PMC4900267 DOI: 10.1074/jbc.m116.726968] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/01/2016] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress arises from an imbalance in the production and scavenging rates of reactive oxygen species (ROS) and is a key factor in the pathophysiology of cardiovascular disease and aging. The presence of parallel pathways and multiple intracellular compartments, each having its own ROS sources and antioxidant enzymes, complicates the determination of the most important regulatory nodes of the redox network. Here we quantified ROS dynamics within specific intracellular compartments in the cytosol and mitochondria and determined which scavenging enzymes exert the most control over antioxidant fluxes in H9c2 cardiac myoblasts. We used novel targeted viral gene transfer vectors expressing redox-sensitive GFP fused to sensor domains to measure H2O2 or oxidized glutathione. Using genetic manipulation in heart-derived H9c2 cells, we explored the contribution of specific antioxidant enzymes to ROS scavenging and glutathione redox potential within each intracellular compartment. Our findings reveal that antioxidant flux is strongly dependent on mitochondrial substrate catabolism, with availability of NADPH as a major rate-controlling step. Moreover, ROS scavenging by mitochondria significantly contributes to cytoplasmic ROS handling. The findings provide fundamental information about the control of ROS scavenging by the redox network and suggest novel interventions for circumventing oxidative stress in cardiac cells.
Collapse
Affiliation(s)
- Swati Dey
- From the Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Agnieszka Sidor
- From the Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Brian O'Rourke
- From the Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
62
|
Effectiveness of N-acetylcysteine for preserving residual renal function in patients undergoing maintenance hemodialysis: multicenter randomized clinical trial. Clin Exp Nephrol 2016; 21:342-349. [PMID: 27206513 DOI: 10.1007/s10157-016-1277-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 05/05/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND To investigate the efficacy and safety of oral N-acetylcysteine (NAC) for preserving residual renal function in patients undergoing hemodialysis. METHODS Randomized, multi-center, parallel-group, open-label clinical trial (Registration No. IRCT 2014071418482N1). 54 patients who have been undergoing hemodialysis for at least 3 months and had residual urine volume >100 ml/24 h were randomly allocated to NAC or no medication. Residual renal function evaluated by (1) estimated glomerular filtration rate (GFR), (2) 24 h urine volume, and (3) renal Kt/V. GFR and Kt/V was determined at baseline and after 3 months. 24 h urine volume was measured at baseline, after 1, 2, and 3 months. RESULTS Intention-to-treat analysis was performed on 47 patients (NAC = 26, control = 21). GFR in patients receiving NAC improved, whereas in the control arm a decline of 1.0 ml/min/1.73 m2 was recorded (3.59 vs. 2.11 ml/min/1.73 m2, effect size = 17.0 %, p = 0.004). For 24 h urine volume, the between-group difference after 1 month was significant (669 vs. 533 ml/24 h, effect size = 15.4 %, p = 0.004). After 3 months, 24 h urine volume in the NAC arm was on average 137 ml higher than in the control group, and the difference reached near significance (673 vs. 536 ml/24 h, p = 0.072). In the follow-up visit, Kt/V was higher in the NAC arm but the difference did not reach statistical significance (0.81 vs. 0.54, p = 0.152). CONCLUSION Three months treatment with NAC appears to be effective in preserving renal function in patients undergoing hemodialysis and the medication is generally well-tolerated.
Collapse
|
63
|
Watanabe Y, Cohen RA, Matsui R. Redox Regulation of Ischemic Angiogenesis - Another Aspect of Reactive Oxygen Species. Circ J 2016; 80:1278-84. [PMID: 27151566 DOI: 10.1253/circj.cj-16-0317] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antioxidants are expected to improve cardiovascular disease (CVD) by eliminating oxidative stress, but clinical trials have not shown promising results in chronic CVD. Animal studies have revealed that reactive oxygen species (ROS) exacerbate acute CVDs in which high levels of ROS are observed. However, ROS are also necessary for angiogenesis after ischemia, because ROS not only damage cells but also stimulate the cell signaling required for angiogenesis. ROS affect signaling by protein modifications, especially of cysteine amino acid thiols. Although there are several cysteine modifications, S-glutathionylation (GSH adducts; -SSG), a reversible cysteine modification by glutathione (GSH), plays an important role in angiogenic signal transduction by ROS. Glutaredoxin-1 (Glrx) is an enzyme that specifically removes GSH adducts in vivo. Overexpression of Glrx inhibits, whereas deletion of Glrx improves revascularization after mouse hindlimb ischemia. These studies indicate that increased levels of GSH adducts in ischemic muscle are beneficial in promoting angiogenesis. The underlying mechanism can be explained by multiple targets of S-gluathionylation, which mediate the angiogenic effects in ischemia. Increments in the master angiogenic transcriptional factor, HIF-1α, reduction of the anti-angiogenic factor sFlt1, activation of the endoplasmic reticulum Ca(2+)pump, SERCA, and inhibition of phosphatases may occur as a consequence of enhanced S-glutathionylation in ischemic tissue. In summary, inducing S-glutathionylation by inhibiting Glrx may be a therapeutic strategy to improve ischemic angiogenesis in CVD. (Circ J 2016; 80: 1278-1284).
Collapse
Affiliation(s)
- Yosuke Watanabe
- Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine
| | | | | |
Collapse
|
64
|
Giam B, Kaye DM, Rajapakse NW. Role of Renal Oxidative Stress in the Pathogenesis of the Cardiorenal Syndrome. Heart Lung Circ 2016; 25:874-80. [PMID: 27132623 DOI: 10.1016/j.hlc.2016.02.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/19/2016] [Accepted: 02/24/2016] [Indexed: 10/24/2022]
Abstract
Renal dysfunction and heart failure commonly co-exist; it is termed the cardiorenal syndrome (CRS). This combination of renal and cardiac impairment presents a substantial clinical challenge and is associated with adverse prognosis. The pathogenesis of the CRS is complex, including chronic activation of the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system, together with reduced renal perfusion. Chronic activation of the RAAS can impair mitochondrial function, and increase mitochondrial derived oxidative stress which in turn can lead to renal injury and sodium and water retention. For example, it has been shown that exogenous Ang II augments renal mitochondrial oxidative stress, reduces GFR and induces albuminuria in rats with heart failure. Administration of Ang II also augmented renal mitochondrial dysfunction in aged mice. Current treatments for CRS, including angiotensin-converting enzyme inhibitors, exert limited renal protection if any at all. Therefore, novel treatments particularly those that can target renal mechanisms downstream to chronic activation of the renal renin-angiotensin system are likely to exert renoprotection in the setting of CRS.
Collapse
Affiliation(s)
- Beverly Giam
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Vic., Australia; Central Clinical School, Monash University, Melbourne, Vic., Australia.
| | - David M Kaye
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Vic., Australia; Department of Medicine, Monash University, Melbourne, Vic., Australia
| | - Niwanthi W Rajapakse
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Vic., Australia; Department of Physiology, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
65
|
Sohrabi Z, Eftekhari MH, Eskandari MH, Rezaianzadeh A, Sagheb MM. Intradialytic Oral Protein Supplementation and Nutritional and Inflammation Outcomes in Hemodialysis: A Randomized Controlled Trial. Am J Kidney Dis 2016; 68:122-30. [PMID: 27086768 DOI: 10.1053/j.ajkd.2016.02.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 02/21/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Malnutrition is a common finding in hemodialysis patients and can increase oxidative stress and inflammation levels. STUDY DESIGN A randomized, controlled, nonblinded, parallel trial. SETTING & PARTICIPANTS 92 hemodialysis patients from a single center with malnutrition according to subjective global assessment (SGA) score (SGA score > 7). INTERVENTION 3 treatment groups (23 patients each) received 220mL of fermented vitamin E-fortified whey beverage (15g of whey protein concentrate + 600IU of vitamin E) or 220mL of fermented whey beverage (15g of whey protein concentrate) or vitamin E (600IU) 3 times a week for 8 weeks. The control group (23 patients) received no intervention. OUTCOME & MEASUREMENTS Primary outcomes were change in SGA score and malnutrition-inflammation score (MIS) from baseline to the end of the trial. RESULTS At the end of the study, 83 patients were analyzed (2, 3, 1, and 3 patients left the study in the vitamin E-fortified whey beverage, whey beverage, vitamin E, and control groups, respectively). Changes in SGA scores were -3.48 (95% CI, -4.90 to -2.00), -3.22 (95% CI, -4.13 to -2.30), -1.70 (95% CI, -3.20 to -0.24), and 1.56 (95% CI, 0.60 to 2.50) for the vitamin E-fortified whey beverage, whey beverage, vitamin E, and control groups, respectively (overall P<0.001; P≤0.001 for each treatment group vs control). Changes in MISs were -3.17 (95% CI, -4.40 to -1.90), -1.83 (95% CI, -2.50 to -1.10), -2.30 (95% CI, -3.50 to -1.10), and 1.48 (95% CI, 0.65 to 2.30) for the vitamin E-fortified whey beverage, whey beverage, vitamin E, and control groups, respectively (overall P<0.001; P<0.001 for each treatment group vs control). Few adverse effects were reported in any group. LIMITATIONS Lack of blinding, small sample size, and short duration. CONCLUSIONS Whey protein in the form of a new fermented whey beverage and vitamin E supplementation may improve SGA score and MIS in the short term.
Collapse
Affiliation(s)
- Zahra Sohrabi
- School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hassan Eftekhari
- Clinical Nutrition Department, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Hadi Eskandari
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Abbas Rezaianzadeh
- Department of Epidemiology, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mahdi Sagheb
- Nephrology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
66
|
Hernández-Reséndiz S, Correa F, García-Niño WR, Buelna-Chontal M, Roldán FJ, Ramírez-Camacho I, Delgado-Toral C, Carbó R, Pedraza-Chaverrí J, Tapia E, Zazueta C. Cardioprotection by curcumin post-treatment in rats with established chronic kidney disease. Cardiovasc Drugs Ther 2016; 29:111-20. [PMID: 25779825 DOI: 10.1007/s10557-015-6581-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE The pathogenic mechanisms leading to cardiovascular disorders in patients with chronic kidney disease have not been clearly established, although increased oxidative stress has been pointed out as a potential cause. Therefore, as cardiovascular events are still the first cause of death in patients with chronic kidney disease and traditional drugs or therapies rarely have effects on cardiac complications, we sought to determine the effect of curcumin in treating cardiac dysfunction in rats with established chronic renal disease. METHODS AND RESULTS Treatment consisted in daily administration of curcumin (120 mg/kg/day) dissolved in 0.05% carboxymethylcellulose via oral gavages during 30 days, beginning from day 30 after 5/6 nephrectomy (5/6Nx). Cardiac function, markers of oxidative stress, activation of PI3K/Akt/GSK3β and MEK1/2-ERK1/2 pathway, metalloproteinase-II (MMP-2) content, overall gelatinolytic activity, ROS production and mitochondrial integrity were evaluated after 1-month treatment. Curcumin restored systolic blood pressure, diminished interventricular and rear wall thickening, decreased left ventricle dimension at end-systole (LVSd) and restored ejection fraction in nephrectomized rats. Also, it diminished metalloproteinase-II levels and overall gelatinase activity, decreased oxidative stress and inhibited the mitochondrial permeability transition pore opening. CONCLUSION Our findings suggest that curcumin might have therapeutic potential in treatment of heart disease in patients with established CKD by attenuating oxidative stress-related events as cardiac remodeling, mitochondrial dysfunction and cell death.
Collapse
Affiliation(s)
- Sauri Hernández-Reséndiz
- Department of Cardiovascular Biomedicine, National Institute of Cardiology I. Ch, Juan Badiano No. 1. Colonia Sección XVI, México, 14080, DF, Mexico
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Li EG, Tian J, Xu ZH. Effects of Gingko biloba extract (EGb 761) on vascular smooth muscle cell calcification induced by β-glycerophosphate. Ren Fail 2016; 38:552-7. [DOI: 10.3109/0886022x.2016.1148724] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
68
|
Pathophysiologic and treatment strategies for cardiovascular disease in end-stage renal disease and kidney transplantations. Cardiol Rev 2016; 23:109-18. [PMID: 25420053 DOI: 10.1097/crd.0000000000000044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The inextricable link between the heart and the kidneys predestines that significant cardiovascular disease ensues in the face of end-stage renal disease (ESRD). As a point of fact, the leading cause of mortality of patients on dialysis is still from cardiovascular etiologies, albeit differing in particular types of disease from the general population. For example, sudden cardiac death outnumbers coronary artery disease in patients with ESRD, which is the reverse for the general population. In this review, we will focus on the pathophysiology and treatment options of important traditional and nontraditional risk factors for cardiovascular disease in ESRD patients such as hypertension, anemia, vascular calcification, hyperparathyroidism, uremia, and oxidative stress. The evidence of erythropoietin-stimulating agents, phosphate binders, calcimimetics, and dialysis modalities will be presented. We will then discuss how these risk factors may be changed and perhaps exacerbated after renal transplantation. This is largely due to the immunosuppressive agents that are both crucial yet potentially detrimental in the posttransplant state. Calcineurin inhibitors, corticosteroids, and mammalian target of rapamycin inhibitors, the mainstay of transplant immunosuppression, are all known to increase the risks of developing new onset diabetes as well as the metabolic syndrome. Thus, we need to carefully negotiate between patients' cardiovascular profile and their risks of rejection. Finally, we end by considering strategies by which we may minimize cardiovascular disease in the transplant population, as this modality still confers the highest chance of survival in patients with ESRD.
Collapse
|
69
|
Machowska A, Carrero JJ, Lindholm B, Stenvinkel P. Therapeutics targeting persistent inflammation in chronic kidney disease. Transl Res 2016; 167:204-13. [PMID: 26173187 DOI: 10.1016/j.trsl.2015.06.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/16/2015] [Accepted: 06/18/2015] [Indexed: 12/24/2022]
Abstract
Systemic inflammation is a condition intrinsically linked to chronic kidney disease (CKD) and its other typical sequelae, such as acquired immune dysfunction, protein-energy wasting (PEW), and accelerated vascular aging that promote premature cardiovascular disease (CVD) and infections, the two leading causes of death in CKD patients. Inflammation is a major contributor to complications in CKD, and inflammatory markers, such as C-reactive protein and pro- and anti-inflammatory cytokines, correlate with underlying causes and consequences of the inflamed uremic phenotype, such as oxidative stress, endothelial dysfunction, CVD, PEW, and infections, and are sensitive and independent predictors of outcome in CKD. Therefore, inflammation appears to be a logical target for potential preventive and therapeutic interventions in patients with CKD. Putative anti-inflammatory therapy strategies aiming at preventing complications and improving outcomes in CKD span over several areas: (1) dealing with the source of inflammation (such as cardiovascular, gastrointestinal or periodontal disease and depression); (2) providing nonspecific immune modulatory effects by promoting healthy dietary habits and other lifestyle changes; (3) promoting increased use of recognized pharmacologic interventions that have pleiotropic effects; and, (4) introducing novel targeted anticytokine interventions. This review provides a brief update on inflammatory biomarkers and possible therapeutic approaches targeting inflammation and the uremic inflammatory milieu in patients with CKD.
Collapse
Affiliation(s)
- Anna Machowska
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Juan Jesus Carrero
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Bengt Lindholm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Stenvinkel
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
70
|
Coombes JS, Sharman JE, Fassett RG. Astaxanthin has no effect on arterial stiffness, oxidative stress, or inflammation in renal transplant recipients: a randomized controlled trial (the XANTHIN trial). Am J Clin Nutr 2016; 103:283-9. [PMID: 26675778 DOI: 10.3945/ajcn.115.115477] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/03/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND There is evidence that renal transplant recipients have accelerated atherosclerosis that is manifest by increased cardiovascular morbidity and mortality. The high incidence of atherosclerosis is, in part, related to increased arterial stiffness, vascular dysfunction, elevated oxidative stress, and inflammation associated with immunosuppressive therapy. The carotenoid astaxanthin has shown potent antioxidant and anti-inflammatory properties. OBJECTIVE The aim was to investigate the effects of oral astaxanthin on arterial stiffness, oxidative stress, and inflammation in renal transplant recipients. DESIGN This trial used a randomized, placebo-controlled, double-blind design in which 61 patients received either 12 mg astaxanthin/d or an identical placebo orally for 1 y. Primary outcomes were 1) arterial stiffness measured by aortic pulse wave velocity (PWV), 2) oxidative stress assessed by total plasma F2-isoprostanes, and 3) inflammation assessed by plasma pentraxin-3. Secondary outcomes included vascular function, carotid artery intima-media thickness, augmentation index, central blood pressure, subendocardial viability ratio, and additional measures of oxidative stress and inflammation. Patients underwent assessments at baseline and at 6 and 12 mo. RESULTS Fifty-eight participants completed the study. There were no significant between-group differences in the changes in any of the primary outcome measures (PWV changed by +9.5% and +6.0%, F2-isoprostanes changed by -3.0% and -9.7%, and pentraxin-3 changed by +50.6% and -11.0% in the placebo and astaxanthin groups, respectively). There were no significant between-group differences in secondary outcome measures. Larger-than-expected variability decreased the power of the study and increased the possibility of a type 2 statistical error. CONCLUSION Astaxanthin (12 mg/d for 12 mo) had no effect on arterial stiffness, oxidative stress, or inflammation in renal transplant recipients. This trial was registered at the Australian New Zealand Clinical Trials Registry (http://www.anzctr.org.au/) as ACTRN12608000159358.
Collapse
Affiliation(s)
- Jeff S Coombes
- School of Human Movement and Nutrition Sciences, University of Queensland, St. Lucia, Australia; and
| | - James E Sharman
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia
| | - Robert G Fassett
- School of Human Movement and Nutrition Sciences, University of Queensland, St. Lucia, Australia; and
| |
Collapse
|
71
|
Abstract
Mechanistic target of rapamycin (mTOR, also known as mammalian target of rapamycin) is a ubiquitous serine/threonine kinase that regulates cell growth, proliferation and survival. These effects are cell-type-specific, and are elicited in response to stimulation by growth factors, hormones and cytokines, as well as to internal and external metabolic cues. Rapamycin was initially developed as an inhibitor of T-cell proliferation and allograft rejection in the organ transplant setting. Subsequently, its molecular target (mTOR) was identified as a component of two interacting complexes, mTORC1 and mTORC2, that regulate T-cell lineage specification and macrophage differentiation. mTORC1 drives the proinflammatory expansion of T helper (TH) type 1, TH17, and CD4(-)CD8(-) (double-negative, DN) T cells. Both mTORC1 and mTORC2 inhibit the development of CD4(+)CD25(+)FoxP3(+) T regulatory (TREG) cells and, indirectly, mTORC2 favours the expansion of T follicular helper (TFH) cells which, similarly to DN T cells, promote B-cell activation and autoantibody production. In contrast to this proinflammatory effect of mTORC2, mTORC1 favours, to some extent, an anti-inflammatory macrophage polarization that is protective against infections and tissue inflammation. Outside the immune system, mTORC1 controls fibroblast proliferation and chondrocyte survival, with implications for tissue fibrosis and osteoarthritis, respectively. Rapamycin (which primarily inhibits mTORC1), ATP-competitive, dual mTORC1/mTORC2 inhibitors and upstream regulators of the mTOR pathway are being developed to treat autoimmune, hyperproliferative and degenerative diseases. In this regard, mTOR blockade promises to increase life expectancy through treatment and prevention of rheumatic diseases.
Collapse
Affiliation(s)
- Andras Perl
- Division of Rheumatology, Departments of Medicine, Microbiology and Immunology, and Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, 750 East Adams Street, Syracuse, New York 13210, USA
| |
Collapse
|
72
|
Granata S, Dalla Gassa A, Tomei P, Lupo A, Zaza G. Mitochondria: a new therapeutic target in chronic kidney disease. Nutr Metab (Lond) 2015; 12:49. [PMID: 26612997 PMCID: PMC4660721 DOI: 10.1186/s12986-015-0044-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/18/2015] [Indexed: 12/24/2022] Open
Abstract
Cellular metabolic changes during chronic kidney disease (CKD) may induce higher production of oxygen radicals that play a significant role in the progression of renal damage and in the onset of important comorbidities. This condition seems to be in part related to dysfunctional mitochondria that cause an increased electron "leakage" from the respiratory chain during oxidative phosphorylation with a consequent generation of reactive oxygen species (ROS). ROS are highly active molecules that may oxidize proteins, lipids and nucleic acids with a consequent damage of cells and tissues. To mitigate this mitochondria-related functional impairment, a variety of agents (including endogenous and food derived antioxidants, natural plants extracts, mitochondria-targeted molecules) combined with conventional therapies could be employed. However, although the anti-oxidant properties of these substances are well known, their use in clinical practice has been only partially investigated. Additionally, for their correct utilization is extremely important to understand their effects, to identify the correct target of intervention and to minimize adverse effects. Therefore, in this manuscript, we reviewed the characteristics of the available mitochondria-targeted anti-oxidant compounds that could be employed routinely in our nephrology, internal medicine and renal transplant centers. Nevertheless, large clinical trials are needed to provide more definitive information about their use and to assess their overall efficacy or toxicity.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Alessandra Dalla Gassa
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Paola Tomei
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126 Verona, VR Italy
| |
Collapse
|
73
|
Hildebrandt W, Sauer R, Bonaterra G, Dugi KA, Edler L, Kinscherf R. Oral N-acetylcysteine reduces plasma homocysteine concentrations regardless of lipid or smoking status. Am J Clin Nutr 2015; 102:1014-24. [PMID: 26447155 DOI: 10.3945/ajcn.114.101964] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 08/27/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Elevated total plasma homocysteine (tHcy) is considered to be an independent cardiovascular disease risk factor, although tHcy lowering by B-vitamins improves only certain clinical endpoints. N-acetylcysteine (NAC), a thiol-containing antioxidant, acutely lowers tHcy and possibly also blood pressure. However, to our knowledge, at present no conclusive long-term evaluation exists that controls for factors such as hyperlipidemia, smoking, medication, and disease stage, all of which affect the thiol redox state, including tHcy. OBJECTIVE We reanalyzed 2 double-blind, placebo-controlled trials in unmedicated middle-aged men, one in a hyperlipidemic group (HYL group; n = 40) and one in a normolipidemic group (NOL group; n = 42), each stratified for smokers and nonsmokers. DESIGN We evaluated the effect of 4 wk of oral NAC (1.8 g/d) on tHcy (primary endpoint), plasma thiol (cysteine), and intracellular glutathione concentrations as well as on blood pressure. The HYL group had total cholesterol >220 mg/dL or triglycerides >150 mg/dL. RESULTS NAC treatment significantly (P = 0.001, multivariate analysis of variance for repeated measures) lowered postabsorptive plasma concentrations of tHcy by -11.7% ± 3.0% (placebo: 4.1% ± 3.6%) while increasing those of cysteine by 28.1% ± 5.7% (placebo: 4.0% ± 3.4%) with no significant impact of hyperlipidemia or smoking. Moreover, NAC significantly decreased systolic (P = 0.003) and diastolic (P = 0.017) blood pressure within all subjects with a significant reduction in diastolic pressure in the HYL group (P = 0.008) but not in the NOL group. An explorative stepwise multiple regression analysis identified 1) post-treatment cysteine as well as 2) pretreatment tHcy and 3) albumin plasma concentrations as being significant contributors to tHcy reduction. CONCLUSIONS Four weeks of oral NAC treatment significantly decreased plasma tHcy concentrations, irrespective of lipid or smoking status, and lowered systolic blood pressure in both normolipidemic and hyperlipidemic men, with significant diastolic blood pressure reductions in the HYL group only. Increased oral intake of cysteine may therefore be considered for primary or secondary prevention of vascular events with regard to the 2 independent risk factors of hyperhomocysteinemia and arterial hypertension.
Collapse
Affiliation(s)
| | - Roland Sauer
- Immunochemistry and Department of Neurology, University Hospital Erlangen, Erlangen, Germany
| | | | - Klaus A Dugi
- Internal Medicine I, University of Heidelberg, Heidelberg, Germany; Departments of
| | - Lutz Edler
- Biostatistics, Deutsches Krebsforschungszentrum, Heidelberg, Germany; and
| | | |
Collapse
|
74
|
Perl A. mTOR activation is a biomarker and a central pathway to autoimmune disorders, cancer, obesity, and aging. Ann N Y Acad Sci 2015; 1346:33-44. [PMID: 25907074 DOI: 10.1111/nyas.12756] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a ubiquitous serine/threonine kinase, which plays pivotal roles in integrating growth signals on a cellular level. To support proliferation and survival under stress, two interacting complexes that harbor mTOR, mTORC1 and mTORC2, promote the transcription of genes involved in carbohydrate metabolism and lipogenesis, enhance protein translation, and inhibit autophagy. Although rapamycin was originally developed as an inhibitor of T cell proliferation for preventing organ transplant rejection, its molecular target, mTOR, has been subsequently identified as a central regulator of metabolic cues that drive lineage specification in the immune system. Owing to oxidative stress, the activation of mTORC1 has emerged as a central pathway for the pathogenesis of systemic lupus erythematosus and other autoimmune diseases. Paradoxically, mTORC1 has also been identified as a mediator of the Warburg effect that allows cell survival under hypoxia. Rapamycin and new classes of mTOR inhibitors are being developed to block not only transplant rejection and autoimmunity but also to treat obesity and various forms of cancer. Through preventing these diseases, personalized mTOR blockade holds promise to extend life span.
Collapse
Affiliation(s)
- Andras Perl
- Division of Rheumatology, Department of Medicine State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York.,Division of Rheumatology, Department of Microbiology and Immunology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York
| |
Collapse
|
75
|
Wu PT, Fitschen PJ, Kistler BM, Jeong JH, Chung HR, Aviram M, Phillips SA, Fernhall B, Wilund KR. Effects of Pomegranate Extract Supplementation on Cardiovascular Risk Factors and Physical Function in Hemodialysis Patients. J Med Food 2015; 18:941-9. [PMID: 25826143 DOI: 10.1089/jmf.2014.0103] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The purpose of this study was to evaluate the effects of oral supplementation with pomegranate extract on cardiovascular risk, physical function, oxidative stress, and inflammation in hemodialysis (HD) patients. Thirty-three HD subjects were randomized to the pomegranate (POM) or placebo (CON) group. Patients in POM ingested a 1000 mg capsule of a purified pomegranate polyphenol extract 7 days/week for 6 months. Individuals in CON ingested a noncaloric placebo capsule using the same protocol. Measurements were conducted at baseline and repeated 6 months following the start of the intervention. Brachial blood pressure (BP) was obtained using an automatic digital BP monitor. Cardiovascular risk was assessed using ultrasound and arterial tonometry. Blood samples were collected for the measurements of circulating markers of inflammation, oxidative stress, and antioxidant capacity. Muscle strength and physical function were assessed by isokinetic dynamometry, a validated shuttle walk test, and a battery of tests to assess functional fitness. Systolic blood pressure and diastolic blood pressure were reduced by 24 ± 13.7 and 10 ± 5.3 mmHg, respectively, in POM (P < .05). However, the BP differences in POM were no longer significant after controlling for baseline BP. The paraoxonase-1 activity increased by 26.6% (P < .05) in POM, compared to no significant change in CON. However, pomegranate supplementation had no effect on other markers of cardiovascular disease risk, inflammation and oxidative stress, or measures of physical function and muscle strength. While pomegranate extract supplementation may reduce BP and increase the antioxidant activity in HD patients, it does not improve other markers of cardiovascular risk, physical function, or muscle strength.
Collapse
Affiliation(s)
- Pei-Tzu Wu
- 1 School of Nursing, University of California-Los Angeles , Los Angeles, California, USA .,3 Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA
| | - Peter J Fitschen
- 2 Division of Nutritional Sciences, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA
| | - Brandon M Kistler
- 3 Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA
| | - Jin Hee Jeong
- 3 Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA
| | - Hae Ryong Chung
- 3 Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA
| | - Michael Aviram
- 4 Technion Rappaport Faculty of Medicine, Rambam Medical Center , Haifa, Israel
| | - Shane A Phillips
- 5 Department of Physical Therapy, University of Illinois at Chicago , Chicago, Illinois, USA
| | - Bo Fernhall
- 6 Department of Kinesiology and Nutrition, University of Illinois at Chicago , Chicago, Illinois, USA
| | - Kenneth R Wilund
- 2 Division of Nutritional Sciences, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA .,3 Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign , Urbana, Illinois, USA
| |
Collapse
|
76
|
McCarty MF, DiNicolantonio JJ. The Molecular Biology and Pathophysiology of Vascular Calcification. Postgrad Med 2015; 126:54-64. [DOI: 10.3810/pgm.2014.03.2740] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
77
|
Xu H, Watanabe M, Qureshi AR, Heimbürger O, Bárány P, Anderstam B, Eriksson M, Stenvinkel P, Lindholm B. Oxidative DNA damage and mortality in hemodialysis and peritoneal dialysis patients. Perit Dial Int 2015; 35:206-15. [PMID: 24584621 PMCID: PMC4406316 DOI: 10.3747/pdi.2013.00259] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/02/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIMS Increased oxidative stress in dialysis patients is thought to contribute to increased mortality; however, confirmatory data are scarce. We analyzed the serum concentration of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a marker of oxidative stress, in relation to mortality in hemodialysis (HD) and peritoneal dialysis (PD) patients. METHODS Serum 8-OHdG, interleukin 6 (IL-6), other biochemical markers, Davies comorbidity score, and protein-energy wasting (PEW) were assessed in 303 prevalent patients treated with HD (n = 220; age: 63 ± 14 years) or PD (n = 83; age: 64 ± 14 years). Mortality was assessed after a median follow-up of 31 months. RESULTS The median (25th - 75th percentile) concentration of 8-OHdG was higher in HD than in PD patients: 1.3 ng/mL (0.9 - 1.8 ng/mL) versus 0.5 ng/mL (0.4 - 0.6 ng/mL), p < 0.001. The HD modality (standard β = 0.57, p < 0.001) and dialysis vintage (standard β = 0.12, p = 0.02) were independent predictors of serum 8-OHdG in a multivariable linear regression model including age, sex, body mass index, dialysis modality (HD or PD), preceding time on dialysis (dialysis vintage), PEW, comorbidity score, IL-6, and use of angiotensin converting-enzyme inhibitors or angiotensin II receptor blockers or statins. During follow-up, 107 patients died. In multivariable Cox regression models including all 303 patients and adjusted for age, sex, body mass index, dialysis modality, dialysis vintage, and comorbidity score, 8-OHdG was significantly associated with all-cause mortality (adjusted hazard ratio: 1.40; 95% confidence limits: 1.05, 1.87 for 1 standard deviation increase of 8-OHdG). In subgroup analyses according to dialysis modality, 8-OHdG was associated with mortality in HD patients but not in PD patients. CONCLUSIONS Oxidative stress as assessed by 8-OHdG is an independent predictor of all-cause mortality in dialysis patients. This association was seen in HD patients, but no such association could be demonstrated for PD patients.
Collapse
Affiliation(s)
- Hong Xu
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Makoto Watanabe
- Division of Nephrology, Department of Medicine, Showa University School of Medicine Tokyo, Tokyo, Japan
| | - Abdul Rashid Qureshi
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Olof Heimbürger
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Bárány
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Björn Anderstam
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Monica Eriksson
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Bengt Lindholm
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
78
|
Bossola M, Tazza L. Wishful Thinking: The Surprisingly Sparse Evidence for a Relationship between Oxidative Stress and Cardiovascular Disease in Hemodialysis Patients. Semin Dial 2015; 28:224-30. [PMID: 25641650 DOI: 10.1111/sdi.12345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The increased frequency of cardiovascular disease observed in hemodialysis patients is secondary to the combination of many traditional (age, male sex, hypertension, smoking, diabetes mellitus, and dyslipidemia) and novel and uremia-related (inflammation, uremic toxins, adipokine imbalance, coagulation disorders, protein-energy wasting, volume overload, endothelial dysfunction, hyperparathyroidism, and subclinical hypothyroidism) risk factors. Usually, in the latter group, oxidative stress is included. However, after decades of research, it remains essentially unknown if oxidative stress has a causative role in the development of cardiovascular disease in long-term hemodialysis patients because adequate longitudinal studies are lacking. Data deriving from cross-sectional studies suggest that biomarkers of oxidative stress are associated with cardiovascular disease prevalence. Conversely, conflicting and inconclusive results have been obtained on the association between oxidative stress and coronary artery calcification, atherosclerosis, and all-cause and cardiovascular disease-related outcome. It is desirable that further studies are conducted on this topic in the near future.
Collapse
Affiliation(s)
- Maurizio Bossola
- Hemodialysis Unit, Division of Transplantation, Catholic University of the Sacred Heart, "Agostino Gemelli" University Hospital, Rome, Italy
| | | |
Collapse
|
79
|
Raghban A, Kirsop J, Tang WHW. Prevention of Heart Failure in Patients with Chronic Kidney Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2015; 9:428. [PMID: 38993263 PMCID: PMC11238633 DOI: 10.1007/s12170-014-0428-z] [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] [Indexed: 10/24/2022]
Abstract
Patients with chronic kidney disease (CKD) have heightened risk of developing heart failure (HF), yet few clinical studies have directly investigated the pathophysiologic underpinnings or therapeutic strategies to prevent HF. A wide range of clinically available cardiac and renal biomarkers can identify at-risk individuals who would benefit from dietary and lifestyle modifications (exercise prescription, smoking cessation), as well as risk factor modification (blood pressure, glucose, and lipid control). Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers have the most consistent data for risk reduction, while other standard HF drugs such as beta-blockers and mineralocorticoid receptor antagonists have promising findings but no large-scale clinical trial evidence for their routine use to prevent the development and progression of HF in this vulnerable population.
Collapse
Affiliation(s)
- Amr Raghban
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
| | - Jennifer Kirsop
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH 44195, USA
| |
Collapse
|
80
|
Rivara MB, Mehrotra R, Linke L, Ruzinski J, Ikizler TA, Himmelfarb J. A pilot randomized crossover trial assessing the safety and short-term effects of pomegranate supplementation in hemodialysis patients. J Ren Nutr 2015; 25:40-9. [PMID: 25218876 PMCID: PMC4282815 DOI: 10.1053/j.jrn.2014.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/08/2014] [Accepted: 07/12/2014] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Oxidative stress and systemic inflammation are highly prevalent in patients undergoing maintenance hemodialysis (MHD) and are linked to excess cardiovascular risk. This study examined whether short-term supplementation with pomegranate juice and extract is safe and well tolerated by MHD patients. The secondary aim was to assess the effect of pomegranate supplementation on oxidative stress, systemic inflammation, monocyte function, and blood pressure. DESIGN Prospective, randomized, crossover, pilot clinical trial (NCT01562340). SETTING The study was conducted from March to October 2012 in outpatient dialysis facilities in the Seattle metropolitan area. SUBJECTS Twenty-four patients undergoing MHD (men, 64%; mean age, 61 ± 14 years) were randomly assigned to receive pomegranate juice or extract during a 4-week intervention period. After a washout period, all patients received the alternative treatment during a second 4-week intervention period. INTERVENTION Patients assigned to receive pomegranate juice received 100 mL of juice before each dialysis session. Patients assigned to receive pomegranate extract were given 1,050 mg of extract daily. MAIN OUTCOME MEASURES The main outcome measures were safety and tolerability of pomegranate juice and extract. Additional secondary outcomes assessed included serum lipids, laboratory biomarkers of inflammation (C-reactive protein and interleukin 6) and oxidative stress (plasma F2 isoprostanes and isofurans), monocyte cytokine production, and predialysis blood pressure. RESULTS Both pomegranate juice and extract were safe and well tolerated by study participants. Over the study period, neither treatment had a significant effect on lipid profiles, plasma C-reactive protein, interleukin 6, F2-isoprostane or isofuran concentrations, predialysis systolic or diastolic blood pressure nor changed the levels of monocyte cytokine production. CONCLUSIONS Both pomegranate juice and extract are safe and well tolerated by patients undergoing MHD but do not influence markers of inflammation or oxidative stress nor affect predialysis blood pressure.
Collapse
Affiliation(s)
- Matthew B Rivara
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington; Kidney Research Institute, Seattle, Washington.
| | - Rajnish Mehrotra
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington; Kidney Research Institute, Seattle, Washington
| | - Lori Linke
- Kidney Research Institute, Seattle, Washington
| | | | - T Alp Ikizler
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington; Kidney Research Institute, Seattle, Washington
| |
Collapse
|
81
|
Rodrigues SD, França KC, Dallin FT, Fujihara CK, Nascimento AJ, Pecoits-Filho R, Nakao LS. N-acetylcysteine as a potential strategy to attenuate the oxidative stress induced by uremic serum in the vascular system. Life Sci 2014; 121:110-6. [PMID: 25500303 DOI: 10.1016/j.lfs.2014.11.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/31/2014] [Accepted: 11/20/2014] [Indexed: 12/11/2022]
Abstract
AIMS Chronic kidney disease (CKD) progression is accompanied by systemic oxidative stress, which contributes to an increase in the risk of cardiovascular diseases (CVDs). N-acetylcysteine (NAC) is among the most studied antioxidants, but its therapeutic benefits in CKD-associated CVDs remain controversial. Here, we investigated whether NAC could inhibit the oxidative stress induced by uremia in vitro and in vivo. MAIN METHODS Endothelial and smooth muscle cells were challenged with human uremic or non-uremic sera, and the effects of a pre-treatment with 2mM NAC were evaluated. Reactive oxygen species (ROS) production, protein oxidation and total glutathione/glutathione disulfide (tGSH/GSSG) ratios were measured. Five-sixths nephrectomized or sham-operated rats were orally treated (in the drinking water) with 60 mg/kg/day NAC or not treated for 53 days. Plasma cysteine/cystine reduction potential Eh(Cyss/2Cys) was determined as a novel marker of the systemic oxidative stress. KEY FINDINGS NAC inhibited all the determined oxidative stress parameters, likely by increasing the tGSH/GSSG ratio, in both cell lines exposed to uremic serum. Orally administered NAC attenuated the systemic oxidative stress in uremic rats. SIGNIFICANCE The present results indicate that NAC, by preventing GSH depletion in vascular cells exposed to uremic serum and by attenuating the systemic oxidative stress during CKD progression, emerges as a potential strategy to prevent the oxidative stress induced by uremic toxicity in the vascular system.
Collapse
Affiliation(s)
- Silvia D Rodrigues
- Departamento de Patologia Básica, Universidade Federal do Paraná, Centro Politécnico, Curitiba 81531-980, Brazil
| | - Karime C França
- Departamento de Patologia Básica, Universidade Federal do Paraná, Centro Politécnico, Curitiba 81531-980, Brazil
| | - Fernando T Dallin
- Departamento de Patologia Básica, Universidade Federal do Paraná, Centro Politécnico, Curitiba 81531-980, Brazil
| | - Clarice K Fujihara
- Laboratório de Fisiopatologia Renal, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Aguinaldo J Nascimento
- Programa de Pós-Graduação em Ciências Farmacêuticas,Universidade Federal do Paraná, Curitiba 80210-170, Brazil
| | - Roberto Pecoits-Filho
- School of Medicine, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, Brazil
| | - Lia S Nakao
- Departamento de Patologia Básica, Universidade Federal do Paraná, Centro Politécnico, Curitiba 81531-980, Brazil.
| |
Collapse
|
82
|
Alani H, Tamimi A, Tamimi N. Cardiovascular co-morbidity in chronic kidney disease: Current knowledge and future research needs. World J Nephrol 2014; 3:156-168. [PMID: 25374809 PMCID: PMC4220348 DOI: 10.5527/wjn.v3.i4.156] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/30/2014] [Accepted: 10/16/2014] [Indexed: 02/05/2023] Open
Abstract
Chronic kidney disease (CKD) is recognised as a health concern globally and leads to high rates of morbidity, mortality and healthcare expenditure. CKD is itself an independent risk factor for unfavorable health outcomes that include cardiovascular disease (CVD). Coronary artery disease is the primary type of CVD in CKD patients and a significant cause of death among renal transplant patients. Traditional and non-traditional risk factors for CVD exist in patients with CKD. Traditional factors include smoking, hypertension, dyslipidemia and diabetes which are highly prevalent in CKD patients. Non-traditional risk factors of CKD are mainly uraemia-specific and increase in prevalence as kidney function declines. Some examples of uraemia-specific risk factors that have been well documented include low levels of haemoglobin, albuminuria, and abnormal bone and mineral metabolism. Therapeutic interventions targeted at more traditional risk factors which contribute to CVD, have not had the desired effect on lowering CVD events and mortality in those suffering with CKD. Future research is warranted to delineate clear evidence to the benefit of modifying non-traditional risk factors.
Collapse
|
83
|
Silswal N, Touchberry CD, Daniel DR, McCarthy DL, Zhang S, Andresen J, Stubbs JR, Wacker MJ. FGF23 directly impairs endothelium-dependent vasorelaxation by increasing superoxide levels and reducing nitric oxide bioavailability. Am J Physiol Endocrinol Metab 2014; 307:E426-36. [PMID: 25053401 PMCID: PMC4154070 DOI: 10.1152/ajpendo.00264.2014] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/15/2014] [Indexed: 12/17/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is secreted primarily by osteocytes and regulates phosphate and vitamin D metabolism. Elevated levels of FGF23 are clinically associated with endothelial dysfunction and arterial stiffness in chronic kidney disease (CKD) patients; however, the direct effects of FGF23 on endothelial function are unknown. We hypothesized that FGF23 directly impairs endothelial vasorelaxation by hindering nitric oxide (NO) bioavailability. We detected expression of all four subtypes of FGF receptors (Fgfr1-4) in male mouse aortas. Exogenous FGF23 (90-9,000 pg/ml) did not induce contraction of aortic rings and did not relax rings precontracted with PGF2α. However, preincubation with FGF23 (9,000 pg/ml) caused a ∼36% inhibition of endothelium-dependent relaxation elicited by acetylcholine (ACh) in precontracted aortic rings, which was prevented by the FGFR antagonist PD166866 (50 nM). Furthermore, in FGF23-pretreated (9,000 pg/ml) aortic rings, we found reductions in NO levels. We also investigated an animal model of CKD (Col4a3(-/-) mice) that displays highly elevated serum FGF23 levels and found they had impaired endothelium-dependent vascular relaxation and reduced nitrate production compared with age-matched wild types. To elucidate a mechanism for the FGF23-induced impairment, we measured superoxide levels in endothelial cells and aortic rings and found that they were increased following FGF23 treatment. Crucially, treatment with the superoxide scavenger tiron reduced superoxide levels and also restored aortic relaxation to ACh. Therefore, our data suggest that FGF23 increases superoxide, inhibits NO bioavailability, and causes endothelial dysfunction in mouse aorta. Together, these data provide evidence that high levels of FGF23 contribute to cardiovascular dysfunction.
Collapse
Affiliation(s)
- Neerupma Silswal
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri; and
| | - Chad D Touchberry
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri; and
| | - Dorothy R Daniel
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri; and
| | - Darla L McCarthy
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri; and
| | - Shiqin Zhang
- The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Jon Andresen
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri; and
| | - Jason R Stubbs
- The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Michael J Wacker
- Muscle Biology Group, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri; and
| |
Collapse
|
84
|
Najafi A, Mojtahedzadeh M, Ahmadi KH, Abdollahi M, Mousavi M, Chelkeba L, Najmeddin F, Ahmadi A. The immunological benefit of higher dose N-acetyl cysteine following mechanical ventilation in critically ill patients. ACTA ACUST UNITED AC 2014; 22:57. [PMID: 25027749 PMCID: PMC4223415 DOI: 10.1186/2008-2231-22-57] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 07/08/2014] [Indexed: 01/26/2023]
Abstract
BACKGROUND Sepsis complication is a major cause of death in multiple trauma critically ill patients. Defensin (cysteine rich anti-microbial peptides), as an important component of immune system, might play an important role in this process. There is also rising data on immunological effects of N-acetyl-cysteine (NAC), a commonly used anti-oxidant in oxidative stress conditions and glutathione (GSH) deficiencies. The aim of the present study was to evaluate the potential beneficial effects of NAC administration on multiple trauma patients with sepsis. METHODS In a prospective, randomized controlled study, 44 multiple trauma critically ill patients who were mechanically ventilated and met the criteria of sepsis and admitted to the intensive care unit (ICU) were randomized into two groups . Control group received all standard ICU therapies and NAC group received intravenous NAC 3 gr every 6 hours for 72 hours in addition to standard therapies. Acute Physiology and Chronic Health Evaluation II (APACHE II) and Sequential Organ Failure Assessment (SOFA) scores, length of ICU stay, ICU mortality were recorded. Levels of serum Immunoglobulin M (IgM), Human β-Defensin 2 (HβD2) and GSH were assessed at baseline and 24, 72, 120 hours after intervention. RESULTS During a period of 13-month screening, 44 patients underwent randomization but 5 patients had to be excluded. 21 patients in NAC group and 18 patients in control group completed the study. For both groups the length of ICU stay, SOFA score and systemic oxygenation were similar. Mortality rate (40% vs. 22% respectively, p = 0.209) and ventilator days (Mean ± SD 19.82 ± 19.55 days vs. 13.82 ± 11.89 days respectively, p = 0.266) were slightly higher for NAC group. IgM and GSH levels were similar between two groups (p = 0.325, 0.125 respectively), HβD2 levels were higher for NAC group (at day 3). CONCLUSION High dose of NAC administration not only did not improve patients' outcome, but also raised the risk of inflammation and was associated with increased serum creatinine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Arezoo Ahmadi
- Department of Anesthesiology and Critical Care Medicine, Sina Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
85
|
High-dialysate-glucose-induced oxidative stress and mitochondrial-mediated apoptosis in human peritoneal mesothelial cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:642793. [PMID: 24891925 PMCID: PMC4026970 DOI: 10.1155/2014/642793] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/30/2014] [Accepted: 04/01/2014] [Indexed: 11/23/2022]
Abstract
Human peritoneal mesothelial cells (HPMCs) are a critical component of the peritoneal membrane and play a pivotal role in dialysis adequacy. Loss of HPMCs can contribute to complications in peritoneal dialysis. Compelling evidence has shown that high-dialysate glucose is a key factor causing functional changes and cell death in HPMCs. We investigated the mechanism of HPMC apoptosis induced by high-dialysate glucose, particularly the role of mitochondria in the maintenance of HPMCs. HPMCs were incubated at glucose concentrations of 5 mM, 84 mM, 138 mM, and 236 mM. Additionally, N-acetylcysteine (NAC) was used as an antioxidant to clarify the mechanism of high-dialysate-glucose-induced apoptosis. Exposing HPMCs to high-dialysate glucose resulted in substantial apoptosis with cytochrome c release, followed by caspase activation and poly(ADP-ribose) polymerase cleavage. High-dialysate glucose induced excessive reactive oxygen species production and lipid peroxidation as well as oxidative damage to DNA. Mitochondrial fragmentation, multiple mitochondrial DNA deletions, and dissipation of the mitochondrial membrane potential were also observed. The mitochondrial dysfunction and cell death were suppressed using NAC. These results indicated that mitochondrial dysfunction is one of the main causes of high-dialysate-glucose-induced HPMC apoptosis.
Collapse
|
86
|
Impellizzeri D, Esposito E, Attley J, Cuzzocrea S. Targeting inflammation: new therapeutic approaches in chronic kidney disease (CKD). Pharmacol Res 2014; 81:91-102. [PMID: 24602801 DOI: 10.1016/j.phrs.2014.02.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/18/2014] [Accepted: 02/24/2014] [Indexed: 01/14/2023]
Abstract
Chronic inflammation and oxidative stress, features that are closely associated with nuclear factor (NF-κB) activation, play a key role in the development and progression of chronic kidney disease (CKD). Several animal models and clinical trials have clearly demonstrated the effectiveness of angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor blocker (ARB) therapy to improve glomerular/tubulointerstitial damage, reduce proteinuria, and decrease CKD progression, but CKD treatment still represents a clinical challenge. Bardoxolone methyl, a first-in-class oral Nrf-2 (nuclear factor erythroid 2-related factor 2) agonist that until recently showed considerable potential for the management of a range of chronic diseases, had been shown to improve kidney function in patients with advanced diabetic nephropathy (DN) with few adverse events in a phase 2 trial, but a large phase 3 study in patients with diabetes and CKD was halted due to emerging toxicity and death in a number of patients. Instead, palmitoylethanolamide (PEA) a member of the fatty acid ethanolamine family, is a novel non-steroidal, kidney friendly anti-inflammatory and anti-fibrotic agent with a well-documented safety profile, that may represent a potential candidate in treating CKD probably by a combination of pharmacological properties, including some activity at the peroxisome proliferator activated receptor alpha (PPAR-α). The aim of this review is to discuss new therapeutic approaches for the treatment of CKD, with particular reference to the outcome of two therapies, bardoxolone methyl and PEA, to improve our understanding of which pharmacological properties are responsible for the anti-inflammatory effects necessary for the effective treatment of renal disease.
Collapse
Affiliation(s)
- Daniela Impellizzeri
- Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, Messina 31-98166, Italy
| | - Emanuela Esposito
- Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, Messina 31-98166, Italy
| | | | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, Messina 31-98166, Italy; Manchester Biomedical Research Centre, Manchester Royal Infirmary, University of Manchester, United Kingdom.
| |
Collapse
|
87
|
Himmelfarb J, Ikizler TA, Ellis C, Wu P, Shintani A, Dalal S, Kaplan M, Chonchol M, Hakim RM. Provision of antioxidant therapy in hemodialysis (PATH): a randomized clinical trial. J Am Soc Nephrol 2013; 25:623-33. [PMID: 24371300 DOI: 10.1681/asn.2013050545] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Increased markers of oxidative stress and acute-phase inflammation are prevalent in patients undergoing maintenance hemodialysis therapy (MHD), and are associated with increased mortality and hospitalization rates and decreased erythropoietin responsiveness. No adequately powered studies have examined the efficacy of antioxidant therapies on markers of inflammation and oxidative stress. We tested the hypothesis that oral antioxidant therapy over 6 months would decrease selected biomarkers of acute-phase inflammation and oxidative stress and improve erythropoietic response in prevalent MHD patients. In total, 353 patients were enrolled in a prospective, placebo-controlled, double-blind clinical trial and randomly assigned to receive a combination of mixed tocopherols (666 IU/d) plus α-lipoic acid (ALA; 600 mg/d) or matching placebos for 6 months (NCT00237718); 238 patients completed the study. High-sensitivity C-reactive protein (hsCRP) and IL-6 concentration were measured as biomarkers of systemic inflammation, and F2 isoprostanes and isofurans were measured as biomarkers of oxidative stress. The groups did not significantly differ at baseline. At 3 and 6 months, the treatment had no significant effect on plasma hsCRP, IL-6, F2 isoprostane, or isofuran concentrations and did not improve the erythropoietic response. No major adverse events were related to the study drug, and both groups had similar mortality and hospitalization rates during the study. In conclusion, the administration of mixed tocopherols and ALA was generally safe and well tolerated, but did not influence biomarkers of inflammation and oxidative stress or the erythropoietic response.
Collapse
Affiliation(s)
- Jonathan Himmelfarb
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, Washington
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
Cardiac events are the major cause of death in hemodialysis patients. Because of the paucity of randomized clinical trials (RCTs) in hemodialysis patients, most cardiovascular therapies in this population are based on observational studies or results extrapolated from studies that excluded hemodialysis patients. However, associations discovered in observational studies do not prove causality, and these studies often report surrogate outcomes rather than clinical end points. Furthermore, interventions that show effectiveness in the general population may have drastically different outcomes and side effect profiles in hemodialysis patients. This review discusses the results of RCTs undertaken recently to evaluate cardiovascular therapies in hemodialysis patients and emphasizes clinically relevant outcomes. Although some interventions have produced similar outcomes in hemodialysis patients and the general population, others have not, suggesting that the management of cardiovascular disease in hemodialysis patients may require strategies that differ from the best practice guidelines applied to general population.
Collapse
Affiliation(s)
- Michael Allon
- Division of Nephrology, University of Alabama, Birmingham, Alabama
| |
Collapse
|
89
|
Abstract
Oxidative stress is increased in systemic lupus erythematosus (SLE), and it contributes to immune system dysregulation, abnormal activation and processing of cell-death signals, autoantibody production and fatal comorbidities. Mitochondrial dysfunction in T cells promotes the release of highly diffusible inflammatory lipid hydroperoxides, which spread oxidative stress to other intracellular organelles and through the bloodstream. Oxidative modification of self antigens triggers autoimmunity, and the degree of such modification of serum proteins shows striking correlation with disease activity and organ damage in SLE. In T cells from patients with SLE and animal models of the disease, glutathione, the main intracellular antioxidant, is depleted and serine/threonine-protein kinase mTOR undergoes redox-dependent activation. In turn, reversal of glutathione depletion by application of its amino acid precursor, N-acetylcysteine, improves disease activity in lupus-prone mice; pilot studies in patients with SLE have yielded positive results that warrant further research. Blocking mTOR activation in T cells could conceivably provide a well-tolerated and inexpensive alternative approach to B-cell blockade and traditional immunosuppressive treatments. Nevertheless, compartmentalized oxidative stress in self-reactive T cells, B cells and phagocytic cells might serve to limit autoimmunity and its inhibition could be detrimental. Antioxidant therapy might also be useful in ameliorating damage caused by other treatments. This Review thus seeks to critically evaluate the complexity of oxidative stress and its relevance to the pathogenesis and treatment of SLE.
Collapse
|
90
|
Maiolino G, Rossitto G, Caielli P, Bisogni V, Rossi GP, Calò LA. The role of oxidized low-density lipoproteins in atherosclerosis: the myths and the facts. Mediators Inflamm 2013; 2013:714653. [PMID: 24222937 PMCID: PMC3816061 DOI: 10.1155/2013/714653] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/28/2013] [Indexed: 02/07/2023] Open
Abstract
The oxidative modification hypothesis of atherosclerosis, which assigns to oxidized low-density lipoproteins (LDLs) a crucial role in atherosclerosis initiation and progression, is still debated. This review examines the role played by oxidized LDLs in atherogenesis taking into account data derived by studies based on molecular and clinical approaches. Experimental data carried out in cellular lines and animal models of atherosclerosis support the proatherogenic role of oxidized LDLs: (a) through chemotactic and proliferating actions on monocytes/macrophages, inciting their transformation into foam cells; (b) through stimulation of smooth muscle cells (SMCs) recruitment and proliferation in the tunica intima; (c) through eliciting endothelial cells, SMCs, and macrophages apoptosis with ensuing necrotic core development. Moreover, most of the experimental data on atherosclerosis-prone animals benefiting from antioxidant treatment points towards a link between oxidative stress and atherosclerosis. The evidence coming from cohort studies demonstrating an association between oxidized LDLs and cardiovascular events, notwithstanding some discrepancies, seems to point towards a role of oxidized LDLs in atherosclerotic plaque development and destabilization. Finally, the results of randomized clinical trials employing antioxidants completed up to date, despite demonstrating no benefits in healthy populations, suggest a benefit in high-risk patients. In conclusion, available data seem to validate the oxidative modification hypothesis of atherosclerosis, although additional proofs are still needed.
Collapse
Affiliation(s)
- Giuseppe Maiolino
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Giacomo Rossitto
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Paola Caielli
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Valeria Bisogni
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Gian Paolo Rossi
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Lorenzo A. Calò
- Department of Medicine (DIMED), Internal Medicine 4, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
91
|
Sung CC, Hsu YC, Chen CC, Lin YF, Wu CC. Oxidative stress and nucleic acid oxidation in patients with chronic kidney disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:301982. [PMID: 24058721 PMCID: PMC3766569 DOI: 10.1155/2013/301982] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/16/2013] [Accepted: 07/22/2013] [Indexed: 12/19/2022]
Abstract
Patients with chronic kidney disease (CKD) have high cardiovascular mortality and morbidity and a high risk for developing malignancy. Excessive oxidative stress is thought to play a major role in elevating these risks by increasing oxidative nucleic acid damage. Oxidative stress results from an imbalance between reactive oxygen/nitrogen species (RONS) production and antioxidant defense mechanisms and can cause vascular and tissue injuries as well as nucleic acid damage in CKD patients. The increased production of RONS, impaired nonenzymatic or enzymatic antioxidant defense mechanisms, and other risk factors including gene polymorphisms, uremic toxins (indoxyl sulfate), deficiency of arylesterase/paraoxonase, hyperhomocysteinemia, dialysis-associated membrane bioincompatibility, and endotoxin in patients with CKD can inhibit normal cell function by damaging cell lipids, arachidonic acid derivatives, carbohydrates, proteins, amino acids, and nucleic acids. Several clinical biomarkers and techniques have been used to detect the antioxidant status and oxidative stress/oxidative nucleic acid damage associated with long-term complications such as inflammation, atherosclerosis, amyloidosis, and malignancy in CKD patients. Antioxidant therapies have been studied to reduce the oxidative stress and nucleic acid oxidation in patients with CKD, including alpha-tocopherol, N-acetylcysteine, ascorbic acid, glutathione, folic acid, bardoxolone methyl, angiotensin-converting enzyme inhibitor, and providing better dialysis strategies. This paper provides an overview of radical production, antioxidant defence, pathogenesis and biomarkers of oxidative stress in patients with CKD, and possible antioxidant therapies.
Collapse
Affiliation(s)
- Chih-Chien Sung
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Cheng-Kung Road, Neihu, Taipei 114, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei 114, Taiwan
| | - Yu-Chuan Hsu
- Division of Neurology, Department of Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan
| | - Chun-Chi Chen
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Cheng-Kung Road, Neihu, Taipei 114, Taiwan
| | - Yuh-Feng Lin
- Division of Nephrology, Department of Medicine, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City 235, Taiwan
- Graduate Institute of Clinical Medical, Taipei Medical University, Taipei 110, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Cheng-Kung Road, Neihu, Taipei 114, Taiwan
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
92
|
Mima A, Matsubara T, Endo S, Murakami T, Hashimoto Y. Use of a polysulfone hemodialysis membrane may prevent recurrent posterior reversible encephalopathy syndrome in a patient undergoing hemodialysis. Int Urol Nephrol 2013; 46:255-60. [PMID: 23568662 DOI: 10.1007/s11255-013-0434-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 03/27/2013] [Indexed: 11/24/2022]
|
93
|
Saito H. Toxico-pharmacological perspective of the Nrf2-Keap1 defense system against oxidative stress in kidney diseases. Biochem Pharmacol 2013; 85:865-72. [DOI: 10.1016/j.bcp.2013.01.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/05/2013] [Accepted: 01/11/2013] [Indexed: 11/25/2022]
|
94
|
Levin A, Lancashire W, Fassett RG. Targets, trends, excesses, and deficiencies: refocusing clinical investigation to improve patient outcomes. Kidney Int 2013; 83:1001-9. [PMID: 23515054 DOI: 10.1038/ki.2013.91] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Clinical trials in nephrology have focused on achieving targets, supplementing deficiencies, and correcting excesses in order to improve patient outcomes. The majority of interventions have failed to demonstrate benefit and some have caused harm. It may be that therapies aiming to 'normalize' parameters may actually disturb evolutionary adaptation, thus causing harm. By refocusing on the physiology of disease, and complexity of adaptation, we may design better trials. We review successful and unsuccessful trials in nephrology and other disciplines and suggest a set of principles by which to design future clinical trials:(1) acknowledge heterogeneity of chronic kidney disease populations and appropriately characterize populations for studies; (2) develop better validated biomarkers (through proteomics, genomics, and metabolomics) to identify responders and nonresponders to interventions; (3) design interventions that mimic physiological processes without collateral detrimental effects; (4) reconsider the status of the randomized-controlled trial as the only 'gold standard' and perform large-scale pragmatic trials comparing current care with the intervention(s) of interest, and (5) broaden nephrology research culture so that the majority of patients are enrolled into observational cohorts and intervention studies, which foster greater knowledge acquisition and dissemination. Improved understanding of pathophysiological mechanisms, in conjunction with more innovative but stringent clinical trial design, will ultimately lead to improved patient outcomes.
Collapse
Affiliation(s)
- Adeera Levin
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | |
Collapse
|
95
|
Weiss N, Papatheodorou L, Morihara N, Hilge R, Ide N. Aged garlic extract restores nitric oxide bioavailability in cultured human endothelial cells even under conditions of homocysteine elevation. JOURNAL OF ETHNOPHARMACOLOGY 2013; 145:162-7. [PMID: 23127645 DOI: 10.1016/j.jep.2012.10.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/05/2012] [Accepted: 10/24/2012] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Supplementation with aged garlic extract (AGE) has been shown to restore impaired endothelium-dependent vasodilator response in subjects with acutely elevated plasma homocysteine (Hcy) levels after an oral methionine load and in patients with chronic coronary artery disease. Moreover, AGE has been shown to inhibit the progression of coronary calcifications in patients with coronary artery disease. The molecular mechanisms, by which AGE preserves endothelial function is unknown. Our objective was to explore whether AGE preserves endothelial nitric oxide (NO) output even under conditions of elevated Hcy levels by preventing oxidative inactivation of the NO synthase cofactor tetrahydrobiopterin. MATERIAL AND METHODS Endothelial (EA.hy 926) cells were incubated with hypoxanthine, aminopterin, thymidine and methionine (HAT/MET) to increase cellular Hcy levels, and with and without AGE. Agonist stimulated NO output was measured using the fluorescent probe DAF-2, and cellular thiol levels (Hcy, cysteine, reduced and oxidized glutathione) and cellular tetrahydrobiopterin levels were measured by high performance liquid chromatography. RESULTS HAT/MET incubation resulted in significantly increased cellular Hcy levels, unaffected by coincubation with AGE. Elevated Hcy went along with significantly decreased NO output (to 34.4 ± 4.4% of control) and levels of tetrahydrobiopterin (from 4.67 ± 2.17 to 2.17 ± 0.97 pmol/mg). Incubation with AGE (5mg/mL) in HAT/MET-treated cells prevented the declines in NO output and tetrahydrobiopterin levels. AGE increased cellular levels of cysteine and total glutathione, and prevented glutathione and tetrahydrobiopterin oxidation induced by elevated Hcy. CONCLUSION Incubation with AGE preserved normal NO output from endothelial cells even under conditions of elevated Hcy levels by increasing cellular thiol antioxidant and prevention of tetrahydrobiopterin oxidation. This suggests that AGE might be useful in the prevention of endothelial dysfunction.
Collapse
Affiliation(s)
- Norbert Weiss
- Center for Vascular Medicine, Section Angiology, University of Munich Medical Center, City Campus, Munich, Germany.
| | | | | | | | | |
Collapse
|
96
|
Rouhi H, Ganji F. Effects of N-acetyl cysteine on serum lipoprotein (a) and proteinuria in type 2 diabetic patients. J Nephropathol 2013; 2:61-6. [PMID: 24475426 DOI: 10.5812/nephropathol.8940] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 05/15/2012] [Accepted: 05/30/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND About 30-40% of diabetic patients who developed nephropathy have lipoprotein disorders, especially lipoprotein a [Lp(a)], which is related to atherosclerosis. OBJECTIVES The aim of this study was to investigate the effect of N-acetyl cysteine (NAC) on the serum levels of Lp(a) and amount of proteinuria in a group of type 2 diabetic patients with diabetic nephropathy. PATIENTS AND METHODS A total of 40, type 2 diabetic (T2D) patients, patients with proteinuria, were randomly divided into two groups. The experimental group was treated by NAC (1200 mg/day) for two months in conjunction with conventional treatment for diabetes and hypertension. Control group received routine medications. RESULTS No significant change was identified in serum Lp(a) during treatment with NAC (P >0.05). However, NAC decreased the amount of proteinuria, serum triglyceride (TG) level and systolic blood pressure in experimental group compared to the control group (P <0.05). CONCLUSIONS These findings suggest that treatment with NAC has no significant effect on the serum level of Lp (a). However, it has beneficial effects on the reduction of proteinuria, serum TG level and systolic blood pressure in T2D patients with nephropathy. Further prospective studies are needed to determine its full role.
Collapse
Affiliation(s)
- Hamid Rouhi
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Forouzan Ganji
- Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
97
|
Kim JC, Kalantar-Zadeh K, Kopple JD. Frailty and protein-energy wasting in elderly patients with end stage kidney disease. J Am Soc Nephrol 2012; 24:337-51. [PMID: 23264684 DOI: 10.1681/asn.2012010047] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Older people constitute an increasingly greater proportion of patients with advanced CKD, including those patients undergoing maintenance dialysis treatment. Frailty is a biologic syndrome of decreased reserve and resistance to stressors that results from cumulative declines across multiple physiologic systems and causes vulnerability to adverse outcomes. Frailty is common in elderly CKD patients, and it may be associated with protein-energy wasting (PEW), sarcopenia, dynapenia, and other complications of CKD. Causes of frailty with or without PEW in the elderly with CKD can be classified into three categories: causes primarily caused by aging per se, advanced CKD per se, or a combination of both conditions. Frailty and PEW in elderly CKD patients are associated with impaired physical performance, disability, poorer quality of life, and reduced survival. Prevention and treatment of these conditions in the elderly CKD patients often require a multifaceted approach. Here, we examine the causes and consequences of these conditions and examine the interplay between frailty and PEW in elderly CKD patients.
Collapse
Affiliation(s)
- Jun Chul Kim
- Division of Nephrology and Hypertension, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | | | | |
Collapse
|
98
|
Jun M, Lv J, Perkovic V, Jardine MJ. Managing cardiovascular risk in people with chronic kidney disease: a review of the evidence from randomized controlled trials. Ther Adv Chronic Dis 2012; 2:265-78. [PMID: 23251754 DOI: 10.1177/2040622311401775] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cardiovascular disease is the leading cause of death and morbidity in people with chronic kidney disease (CKD) making measures to modify cardiovascular risk a clinical priority. The relationship between risk factors and cardiovascular outcomes is often substantially different in people with CKD compared with the general population, leading to uncertainty around pathophysiological mechanisms and the validity of generalizations from the general population. Furthermore, published reports of subgroup analyses from clinical trials have suggested that a range of interventions may have different effects in people with kidney disease compared with those with normal kidney function. There is a relative scarcity of randomized controlled trials (RCTs) conducted in CKD populations, and most such trials are small and underpowered. As a result, evidence to support cardiovascular risk modification measures for people with CKD is largely derived from small trials and post hoc analyses of RCTs conducted in the general population. In this review, we examine the available RCT evidence on interventions aimed at preventing cardiovascular events in people with kidney disease to identify beneficial treatments as well as current gaps in knowledge that should be a priority for future research.
Collapse
Affiliation(s)
- Min Jun
- The George Institute for Global Health, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
99
|
Camuglia AC, Maeder MT, Starr J, Farrington C, Kaye DM. Impact of N-acetylcysteine on endothelial function, B-type natriuretic peptide and renal function in patients with the cardiorenal syndrome: a pilot cross over randomised controlled trial. Heart Lung Circ 2012; 22:256-9. [PMID: 23219310 DOI: 10.1016/j.hlc.2012.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 10/24/2012] [Accepted: 10/27/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Both heart and renal failure are characterised by increased systemic oxidative stress and endothelial dysfunction and occur in the cardiorenal syndrome (CRS). The aim of the present study was to assess the impact of N-acetylcysteine (NAC), a potent antioxidant, on endothelial function, B-type natriuretic peptide (BNP) and renal function in patients with CRS. METHODS In a double blind, placebo controlled manner, we randomised nine stable outpatients with both heart failure (LVEF<40% and NYHA class II or III) and renal failure (Cockroft Gault clearance of 20-60ml/min) to placebo or NAC (500mg orally twice daily) for 28 days followed by a wash out period (>7 days) and crossover to the other treatment. RESULTS Eight patients completed the study and all data (N=9) was used in the analysis. Mean forearm blood flow improved significantly with NAC with mean ratio of improvement of 1.99 (SEM: ±0.49) for NAC and 0.73 (SEM: ±0.23) for placebo with a p-value of 0.047. There was no significant difference in BNP (p=0.25), renal function (p=0.71) or NYHA class (p=0.5). No deaths occurred during the trial. CONCLUSION In this pilot trial of patients with CRS, NAC therapy was associated with improved forearm blood flow. This may represent a general improvement in endothelial function and warrants further investigation of antioxidant therapy in these patients.
Collapse
Affiliation(s)
- Anthony C Camuglia
- The Alfred Hospital, Commercial Road, Melbourne 8008, Victoria, Australia
| | | | | | | | | |
Collapse
|
100
|
Feldman L, Abu Hamad R, Efrati S, Ashker A, Beberashvili I, Shani M. Effect of N-acetylcysteine on residual renal function in chronic haemodialysis patients treated with high-flux synthetic dialysis membranes: a pilot study. ISRN NEPHROLOGY 2012; 2013:636208. [PMID: 24977134 PMCID: PMC4045415 DOI: 10.5402/2013/636208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 10/23/2012] [Indexed: 11/23/2022]
Abstract
Background. Preservation of residual renal function in chronic dialysis patients has proven to be a major predictor of survival. The aim of the present study was to investigate an ability of the combined use of N-acetylcysteine and high-flux biocompatible haemodialysis membranes to improve residual renal function in haemodialysis patients. Patients and Methods. Chronic haemodialysis patients with a residual urine output of at least 100 mL/24 h were administered oral an N-acetylcysteine 1200 mg twice daily for 2 weeks. Treatment group included patients treated with dialysers using high-flux synthetic biocompatible membranes. Control group included patients treated with dialysers using low-flux semisyntetic triacetate haemodialysis membranes. Results. Eighteen patients participated in the study. The residual glomerular filtration rate showed a nonsignificant trend for increase in both groups. The magnitude of GFR improvement after N-acetylcysteine administration was less pronounced in the group treated with high-flux biocompatible membranes: +0.17 ± 0.56 mL/min/1.73 m(2) in treatment group and +0.65 ± 0.53 mL/min/1.73 m(2) in control group (P < 0.05). Conclusion. In this study of favorable effect of N-acetylcysteine on residual renal function in chronic haemodialysis patients may be less pronounced when using high-flux biocompatible, rather than low-flux semisyntetic, HD membranes.
Collapse
Affiliation(s)
- Leonid Feldman
- Nephrology Division and Research & Development Unit, Assaf Harofeh Medical Center, Zerifin 70300, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ramzia Abu Hamad
- Nephrology Division and Research & Development Unit, Assaf Harofeh Medical Center, Zerifin 70300, Israel
| | - Shai Efrati
- Nephrology Division and Research & Development Unit, Assaf Harofeh Medical Center, Zerifin 70300, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ali Ashker
- Nephrology Division and Research & Development Unit, Assaf Harofeh Medical Center, Zerifin 70300, Israel
| | - Ilia Beberashvili
- Nephrology Division and Research & Development Unit, Assaf Harofeh Medical Center, Zerifin 70300, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Michal Shani
- Family Medicine Department, Clalit Health Services and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 97904, Israel
| |
Collapse
|