1
|
Hung Y, Cheng CC, Lu YY, Huang SY, Chen YC, Lin FJ, Lin WS, Kao YH, Lin YK, Chen SA, Chen YJ. Indoxyl Sulfate Induces Ventricular Arrhythmias Attenuated by Secretoneurin in Right Ventricular Outflow Tract Cardiomyocytes. Cardiovasc Toxicol 2025; 25:471-485. [PMID: 39838186 DOI: 10.1007/s12012-025-09963-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
Ventricular arrhythmias (VAs) are major causes of sudden cardiac death in chronic kidney disease (CKD) patients. Indoxyl sulfate (IS) is one common uremic toxin found in CKD patients. This study investigated whether IS could induce VAs via increasing right ventricular outflow tract (RVOT) arrhythmogenesis. Using conventional microelectrodes and whole-cell patch clamps, we studied the action potentials (APs) and ionic currents of isolated rabbit RVOT tissue preparations and single cardiomyocytes before and after IS (0.1 and 1.0 μM). Calcium fluorescence imaging was performed in RVOT cardiomyocytes treated with and without IS (1.0 μM) to evaluate the calcium transient and the calcium leak. In rabbit RVOT tissues, IS (0.1 and 1.0 μM) attenuated the contractility and shortened the AP durations in a dose-dependent manner. In addition, IS (0.1 and 1.0 μM) enhanced the pro-arrhythmia effects of isoproterenol (ISO, 1.0 μM) and rapid ventricular pacing in RVOT (before versus after ISO, 25% versus 83%, N = 12). In RVOT cardiomyocytes, IS (1.0 μM) significantly decreased the L-type calcium currents but increased the sodium-calcium exchanger and sodium window currents. Cardiomyocytes treated with IS (1.0 μM) had lower calcium transients but higher diastolic calcium and calcium leak than those without IS treatment. Pretreatment with secretoneurin (SN, 30 nM, a potent neuropeptide, suppressing CaMKII) or KN-93 (0.1 μM, a CaMKII inhibitor) prevented IS-induced ionic current changes and arrhythmogenesis. In conclusion, IS modulates RVOT electrophysiology and arrhythmogenesis via enhanced CaMKII activity, which is attenuated by SN, leading to a novel therapeutic target for CKD arrhythmias.
Collapse
Affiliation(s)
- Yuan Hung
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chen-Chuan Cheng
- Department of Cardiology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Shih-Yu Huang
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Division of Cardiac Electrophysiology, Cardiovascular Center, Cathay General Hospital, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan.
| | - Fong-Jhih Lin
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Shiang Lin
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, No. 250, Wuxing St., Taipei, 11031, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, No. 250, Wuxing St., Taipei, 11031, Taiwan.
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Dimitrios K, Evangelia G, Christodoulos P, Vassilios V. Accelerated idioventricular rhythm as a manifestation of chronic renocardiac syndrome: A case report. Ann Noninvasive Electrocardiol 2024; 29:e13131. [PMID: 38923781 PMCID: PMC11199323 DOI: 10.1111/anec.13131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
In this case report, we describe a patient who presented with chronic symptoms and signs of uremia and persistent accelerated idioventricular rhythm (AIVR) on electrocardiogram. Findings from blood tests, echocardiography, renal ultrasound, and renal scan were suggestive of heart failure with reduced ejection fraction and chronic kidney disease, and attendance of daily hemodialysis sessions led to the restoration of sinus rhythm. Typically, AIVR has a favorable prognosis and, if necessary, medical intervention focuses on addressing the underlying responsible causes. Accumulation of uremic toxins has the potential to trigger the formation of AIVR and clearance of small solutes through conventional hemodialysis may contribute to sinus rhythm restoration.
Collapse
Affiliation(s)
- Kotzadamis Dimitrios
- Third Department of CardiologyHippokration General Hospital of ThessalonikiThessalonikiGreece
| | | | | | - Vassilikos Vassilios
- Third Department of CardiologyHippokration General Hospital of ThessalonikiThessalonikiGreece
| |
Collapse
|
3
|
Soomro QH, Charytan DM. New Insights on Cardiac Arrhythmias in Patients With Kidney Disease. Semin Nephrol 2024; 44:151518. [PMID: 38772780 DOI: 10.1016/j.semnephrol.2024.151518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
The risk of arrhythmia and its management become increasingly complex as kidney disease progresses. This presents a multifaceted clinical challenge. Our discussion addresses these specific challenges relevant to patients as their kidney disease advances. We highlight numerous opportunities for enhancing the current standard of care within this realm. Additionally, this review delves into research concerning early detection, prevention, diagnosis, and treatment of various arrhythmias spanning the spectrum of kidney disease.
Collapse
|
4
|
Biruete A, Chen NX, Metzger CE, Srinivasan S, O'Neill K, Fallen PB, Fonseca A, Wilson HE, de Loor H, Evenepoel P, Swanson KS, Allen MR, Moe SM. The Dietary Fiber Inulin Slows Progression of Chronic Kidney Disease-Mineral Bone Disorder (CKD-MBD) in a Rat Model of CKD. JBMR Plus 2023; 7:e10837. [PMID: 38130753 PMCID: PMC10731114 DOI: 10.1002/jbm4.10837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 12/23/2023] Open
Abstract
Chronic kidney disease (CKD)-mineral bone disorder (CKD-MBD) leads to fractures and cardiovascular disease. Observational studies suggest beneficial effects of dietary fiber on both bone and cardiovascular outcomes, but the effect of fiber on CKD-MBD is unknown. To determine the effect of fiber on CKD-MBD, we fed the Cy/+ rat with progressive CKD a casein-based diet of 0.7% phosphate with 10% inulin (fermentable fiber) or cellulose (non-fermentable fiber) from 22 weeks to either 30 or 32 weeks of age (~30% and ~15% of normal kidney function; CKD 4 and 5). We assessed CKD-MBD end points of biochemistry, bone quantity and quality, cardiovascular health, and cecal microbiota and serum gut-derived uremic toxins. Results were analyzed by two-way analysis of variance (ANOVA) to evaluate the main effects of CKD stage and inulin, and their interaction. The results showed that in CKD animals, inulin did not alter kidney function but reduced the increase from stage 4 to 5 in serum levels of phosphate and parathyroid hormone, but not fibroblast growth factor-23 (FGF23). Bone turnover and cortical bone parameters were similarly improved but mechanical properties were not altered. Inulin slowed progression of aorta and cardiac calcification, left ventricular mass index, and fibrosis. To understand the mechanism, we assessed intestinal microbiota and found changes in alpha and beta diversity and significant changes in several taxa with inulin, together with a reduction in circulating gut derived uremic toxins such as indoxyl sulfate and short-chain fatty acids. In conclusion, the addition of the fermentable fiber inulin to the diet of CKD rats led to a slowed progression of CKD-MBD without affecting kidney function, likely mediated by changes in the gut microbiota composition and lowered gut-derived uremic toxins. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Annabel Biruete
- Department of Nutrition SciencePurdue UniversityWest LafayetteINUSA
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Neal X. Chen
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Corinne E. Metzger
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Shruthi Srinivasan
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Kalisha O'Neill
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Paul B. Fallen
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Austin Fonseca
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Hannah E. Wilson
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Henriette de Loor
- KU Leuven Department of Microbiology and ImmunologyNephrology and Renal Transplantation Research Group, KU LeuvenLeuvenBelgium
| | - Pieter Evenepoel
- KU Leuven Department of Microbiology and ImmunologyNephrology and Renal Transplantation Research Group, KU LeuvenLeuvenBelgium
- Department of Nephrology and Renal TransplantationUniversity Hospitals LeuvenLeuvenBelgium
| | - Kelly S. Swanson
- Department of Animal SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Matthew R. Allen
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Sharon M. Moe
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
5
|
Lu YY, Chen YC, Lin YK, Chen SA, Chen YJ. Electrical and Structural Insights into Right Ventricular Outflow Tract Arrhythmogenesis. Int J Mol Sci 2023; 24:11795. [PMID: 37511554 PMCID: PMC10380666 DOI: 10.3390/ijms241411795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/08/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
The right ventricular outflow tract (RVOT) is the major origin of ventricular arrhythmias, including premature ventricular contractions, idiopathic ventricular arrhythmias, Brugada syndrome, torsade de pointes, long QT syndrome, and arrhythmogenic right ventricular cardiomyopathy. The RVOT has distinct developmental origins and cellular characteristics and a complex myocardial architecture with high shear wall stress, which may lead to its high vulnerability to arrhythmogenesis. RVOT myocytes are vulnerable to intracellular sodium and calcium overload due to calcium handling protein modulation, enhanced CaMKII activity, ryanodine receptor phosphorylation, and a higher cAMP level activated by predisposing factors or pathological conditions. A reduction in Cx43 and Scn5a expression may lead to electrical uncoupling in RVOT. The purpose of this review is to update the current understanding of the cellular and molecular mechanisms of RVOT arrhythmogenesis.
Collapse
Affiliation(s)
- Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24257, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Cardiovacular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Cardiovacular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11696, Taiwan
| |
Collapse
|
6
|
Biruete A, Chen NX, Metzger CE, Srinivasan S, O’Neill K, Fallen PB, Fonseca A, Wilson HE, de Loor H, Evenepoel P, Swanson KS, Allen MR, Moe SM. The Dietary Fermentable Fiber Inulin Alters the Intestinal Microbiome and Improves Chronic Kidney Disease Mineral-Bone Disorder in a Rat Model of CKD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.29.526093. [PMID: 36778372 PMCID: PMC9915522 DOI: 10.1101/2023.01.29.526093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background Dietary fiber is important for a healthy diet, but intake is low in CKD patients and the impact this has on the manifestations of CKD-Mineral Bone Disorder (MBD) is unknown. Methods The Cy/+ rat with progressive CKD was fed a casein-based diet of 0.7% phosphate with 10% inulin (fermentable fiber) or cellulose (non-fermentable fiber) from 22 weeks to either 30 or 32 weeks of age (~30 and ~15 % of normal kidney function). We assessed CKD-MBD, cecal microbiota, and serum gut-derived uremic toxins. Two-way ANOVA was used to evaluate the effect of age and inulin diet, and their interaction. Results In CKD animals, dietary inulin led to changes in microbiota alpha and beta diversity at 30 and 32 weeks, with higher relative abundance of several taxa, including Bifidobacterium and Bacteroides , and lower Lactobacillus . Inulin reduced serum levels of gut-derived uremic toxins, phosphate, and parathyroid hormone, but not fibroblast growth factor-23. Dietary inulin decreased aorta and cardiac calcification and reduced left ventricular mass index and cardiac fibrosis. Bone turnover and cortical bone parameters were improved with inulin; however, bone mechanical properties were not altered. Conclusions The addition of the fermentable fiber inulin to the diet of CKD rats led to changes in the gut microbiota composition, lowered gut-derived uremic toxins, and improved most parameters of CKD-MBD. Future studies should assess this fiber as an additive therapy to other pharmacologic and diet interventions in CKD. Significance Statement Dietary fiber has well established beneficial health effects. However, the impact of fermentable dietary fiber on the intestinal microbiome and CKD-MBD is poorly understood. We used an animal model of progressive CKD and demonstrated that the addition of 10% of the fermentable fiber inulin to the diet altered the intestinal microbiota and lowered circulating gut-derived uremic toxins, phosphorus, and parathyroid hormone. These changes were associated with improved cortical bone parameters, lower vascular calcification, and reduced cardiac hypertrophy, fibrosis and calcification. Taken together, dietary fermentable fiber may be a novel additive intervention to traditional therapies of CKD-MBD.
Collapse
|
7
|
King BMN, Mintz S, Lin X, Morley GE, Schlamp F, Khodadadi-Jamayran A, Fishman GI. Chronic Kidney Disease Induces Proarrhythmic Remodeling. Circ Arrhythm Electrophysiol 2023; 16:e011466. [PMID: 36595632 PMCID: PMC9852080 DOI: 10.1161/circep.122.011466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) are at increased risk of developing cardiac arrhythmogenesis and sudden cardiac death; however, the basis for this association is incompletely known. METHODS Here, using murine models of CKD, we examined interactions between kidney disease progression and structural, electrophysiological, and molecular cardiac remodeling. RESULTS C57BL/6 mice with adenine supplemented in their diet developed progressive CKD. Electrocardiographically, CKD mice developed significant QT prolongation and episodes of bradycardia. Optical mapping of isolated-perfused hearts using voltage-sensitive dyes revealed significant prolongation of action potential duration with no change in epicardial conduction velocity. Patch-clamp studies of isolated ventricular cardiomyocytes revealed changes in sodium and potassium currents consistent with action potential duration prolongation. Global transcriptional profiling identified dysregulated expression of cellular stress response proteins RBM3 (RNA-binding motif protein 3) and CIRP (cold-inducible RNA-binding protein) that may underlay the ion channel remodeling. Unexpectedly, we found that female sex is a protective factor in the progression of CKD and its cardiac sequelae. CONCLUSIONS Our data provide novel insights into the association between CKD and pathologic proarrhythmic cardiac remodeling. Cardiac cellular stress response pathways represent potential targets for pharmacologic intervention for CKD-induced heart rhythm disorders.
Collapse
Affiliation(s)
- Benjamin M N King
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Shana Mintz
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Xianming Lin
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Gregory E Morley
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Florencia Schlamp
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | | | - Glenn I Fishman
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| |
Collapse
|
8
|
van Ham WB, Cornelissen CM, van Veen TAB. Uremic toxins in chronic kidney disease highlight a fundamental gap in understanding their detrimental effects on cardiac electrophysiology and arrhythmogenesis. Acta Physiol (Oxf) 2022; 236:e13888. [PMID: 36148604 PMCID: PMC9787632 DOI: 10.1111/apha.13888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 01/29/2023]
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) have an estimated 700-800 and 523 million cases worldwide, respectively, with CVD being the leading cause of death in CKD patients. The pathophysiological interplay between the heart and kidneys is defined as the cardiorenal syndrome (CRS), in which worsening of kidney function is represented by increased plasma concentrations of uremic toxins (UTs), culminating in dialysis patients. As there is a high incidence of CVD in CKD patients, accompanied by arrhythmias and sudden cardiac death, knowledge on electrophysiological remodeling would be instrumental for understanding the CRS. While the interplay between both organs is clearly of importance in CRS, the involvement of UTs in pro-arrhythmic remodeling is only poorly investigated, especially regarding the mechanistic background. Currently, the clinical approach against potential arrhythmic events is mainly restricted to symptom treatment, stressing the need for fundamental research on UT in relation to electrophysiology. This review addresses the existing knowledge of UTs and cardiac electrophysiology, and the experimental research gap between fundamental research and clinical research of the CRS. Clinically, mainly absorbents like ibuprofen and AST-120 are studied, which show limited safe and efficient usability. Experimental research shows disturbances in cardiac electrical activation and conduction after inducing CKD or exposure to UTs, but are scarcely present or focus solely on already well-investigated UTs. Based on UTs data derived from CKD patient cohort studies, a clinically relevant overview of physiological and pathological UTs concentrations is created. Using this, future experimental research is stimulated to involve electrophysiologically translatable animals, such as rabbits, or in vitro engineered heart tissues.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Carlijn M. Cornelissen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Toon A. B. van Veen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
9
|
Biruete A, Metzger CE, Chen NX, Swallow EA, Vrabec C, Clinkenbeard EL, Stacy AJ, Srinivasan S, O'Neill K, Avin KG, Allen MR, Moe SM. Effects of ferric citrate and intravenous iron sucrose on markers of mineral, bone, and iron homeostasis in a rat model of CKD-MBD. Nephrol Dial Transplant 2022; 37:1857-1867. [PMID: 35482713 PMCID: PMC9494145 DOI: 10.1093/ndt/gfac162] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Anemia and chronic kidney disease-mineral and bone disorder (CKD-MBD) are common and begin early in CKD. Limited studies have concurrently compared the effects of ferric citrate (FC) versus intravenous (IV) iron on CKD-MBD and iron homeostasis in moderate CKD. METHODS We tested the effects of 10 weeks of 2% FC versus IV iron sucrose in rats with moderate CKD (Cy/+ male rat) and untreated normal (NL) littermates. Outcomes included a comprehensive assessment of CKD-MBD, iron homeostasis and oxidative stress. RESULTS CKD rats had azotemia, elevated phosphorus, parathyroid hormone and fibroblast growth factor-23 (FGF23). Compared with untreated CKD rats, treatment with FC led to lower plasma phosphorus, intact FGF23 and a trend (P = 0.07) toward lower C-terminal FGF23. FC and IV iron equally reduced aorta and heart calcifications to levels similar to NL animals. Compared with NL animals, CKD animals had higher bone turnover, lower trabecular volume and no difference in mineralization; these were unaffected by either iron treatment. Rats treated with IV iron had cortical and bone mechanical properties similar to NL animals. FC increased the transferrin saturation rate compared with untreated CKD and NL rats. Neither iron treatment increased oxidative stress above that of untreated CKD. CONCLUSIONS Oral FC improved phosphorus homeostasis, some iron-related parameters and the production and cleavage of FGF23. The intermittent effect of low-dose IV iron sucrose on cardiovascular calcification and bone should be further explored in moderate-advanced CKD.
Collapse
Affiliation(s)
- Annabel Biruete
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Corinne E Metzger
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Neal X Chen
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth A Swallow
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Curtis Vrabec
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Erica L Clinkenbeard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander J Stacy
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shruthi Srinivasan
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kalisha O'Neill
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith G Avin
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indiana University, Indianapolis, IN, USA
| | - Matthew R Allen
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, IN, USA
| | - Sharon M Moe
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, IN, USA
| |
Collapse
|
10
|
Okada Y, Komukai S, Kitamura T, Kiguchi T, Irisawa T, Yamada T, Yoshiya K, Park C, Nishimura T, Ishibe T, Yagi Y, Kishimoto M, Inoue T, Hayashi Y, Sogabe T, Morooka T, Sakamoto H, Suzuki K, Nakamura F, Matsuyama T, Nishioka N, Kobayashi D, Matsui S, Hirayama A, Yoshimura S, Kimata S, Shimazu T, Ohtsuru S, Iwami T. Clinical Phenotyping of Out-of-Hospital Cardiac Arrest Patients With Shockable Rhythm - Machine Learning-Based Unsupervised Cluster Analysis. Circ J 2022; 86:668-676. [PMID: 34732587 DOI: 10.1253/circj.cj-21-0675] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The hypothesis of this study is that latent class analysis could identify the subphenotypes of out-of-hospital cardiac arrest (OHCA) patients associated with the outcomes and allow us to explore heterogeneity in the effects of extracorporeal cardiopulmonary resuscitation (ECPR). METHODS AND RESULTS This study was a retrospective analysis of a multicenter prospective observational study (CRITICAL study) of OHCA patients. It included adult OHCA patients with initial shockable rhythm. Patients from 2012 to 2016 (development dataset) were included in the latent class analysis, and those from 2017 (validation dataset) were included for evaluation. The association between subphenotypes and outcomes was investigated. Further, the heterogeneity of the association between ECPR implementation and outcomes was explored. In the study results, a total of 920 patients were included for latent class analysis. Three subphenotypes (Groups 1, 2, and 3) were identified, mainly characterized by the distribution of partial pressure of O2(PO2), partial pressure of CO2(PCO2) value of blood gas assessment, cardiac rhythm on hospital arrival, and estimated glomerular filtration rate. The 30-day survival outcomes were varied across the groups: 15.7% in Group 1; 30.7% in Group 2; and 85.9% in Group 3. Further, the association between ECPR and 30-day survival outcomes by subphenotype groups in the development dataset was as varied. These results were validated using the validation dataset. CONCLUSIONS The latent class analysis identified 3 subphenotypes with different survival outcomes and potential heterogeneity in the effects of ECPR.
Collapse
Affiliation(s)
- Yohei Okada
- Department of Preventive Services, School of Public Health, Kyoto University
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University
| | - Sho Komukai
- Division of Biomedical Statistics, Department of Integrated Medicine, Graduate School of Medicine, Osaka University
| | - Tetsuhisa Kitamura
- Division of Environmental Medicine and Population Sciences, Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University
| | | | - Taro Irisawa
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University
| | - Tomoki Yamada
- Emergency and Critical Care Medical Center, Osaka Police Hospital
| | - Kazuhisa Yoshiya
- Department of Emergency and Critical Care Medicine, Kansai Medical University, Takii Hospital
| | - Changhwi Park
- Department of Emergency Medicine, Tane General Hospital
| | | | - Takuya Ishibe
- Department of Emergency and Critical Care Medicine, Kindai University Faculty of Medicine
| | | | | | | | | | - Taku Sogabe
- Traumatology and Critical Care Medical Center, National Hospital Organization Osaka National Hospital
| | - Takaya Morooka
- Emergency and Critical Care Medical Center, Osaka City General Hospital
| | | | - Keitaro Suzuki
- Emergency and Critical Care Medical Center, Kishiwada Tokushukai Hospital
| | - Fumiko Nakamura
- Department of Emergency and Critical Care Medicine, Kansai Medical University
| | - Tasuku Matsuyama
- Department of Emergency Medicine, Kyoto Prefectural University of Medicine
| | - Norihiro Nishioka
- Department of Preventive Services, School of Public Health, Kyoto University
| | - Daisuke Kobayashi
- Department of Preventive Services, School of Public Health, Kyoto University
| | - Satoshi Matsui
- Division of Environmental Medicine and Population Sciences, Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University
| | - Atsushi Hirayama
- Public Health, Department of Social and Environmental Medicine, Graduate School of Medicine, Osaka University
| | - Satoshi Yoshimura
- Department of Preventive Services, School of Public Health, Kyoto University
| | - Shunsuke Kimata
- Department of Preventive Services, School of Public Health, Kyoto University
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University
| | - Shigeru Ohtsuru
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University
| | - Taku Iwami
- Department of Preventive Services, School of Public Health, Kyoto University
| |
Collapse
|
11
|
Huang SY, Chen YC, Kao YH, Lu YY, Lin YK, Higa S, Chen SA, Chen YJ. Calcium dysregulation increases right ventricular outflow tract arrhythmogenesis in rabbit model of chronic kidney disease. J Cell Mol Med 2021; 25:11264-11277. [PMID: 34761510 PMCID: PMC8650029 DOI: 10.1111/jcmm.17052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 12/03/2022] Open
Abstract
Chronic kidney disease (CKD) increases the risk of arrhythmia. The right ventricular outflow tract (RVOT) is a crucial site of ventricular tachycardia (VT) origination. We hypothesize that CKD increases RVOT arrhythmogenesis through its effects on calcium dysregulation. We analysed measurements obtained using conventional microelectrodes, patch clamp, confocal microscopy, western blotting, immunohistochemical examination and lipid peroxidation for both control and CKD (induced by 150 mg/kg neomycin and 500 mg/kg cefazolin daily) rabbit RVOT tissues or cardiomyocytes. The RVOT of CKD rabbits exhibited a short action potential duration, high incidence of tachypacing (20 Hz)‐induced sustained VT, and long duration of isoproterenol and tachypacing‐induced sustained and non‐sustained VT. Tachypacing‐induced sustained and non‐sustained VT in isoproterenol‐treated CKD RVOT tissues were attenuated by KB‐R7943 and partially inhibited by KN93 and H89. The CKD RVOT myocytes had high levels of phosphorylated CaMKII and PKA, and an increased expression of tyrosine hydroxylase‐positive neural density. The CKD RVOT myocytes exhibited large levels of Ito, IKr, NCX and L‐type calcium currents, calcium leak and malondialdehyde but low sodium current, SERCA2a activity and SR calcium content. The RVOT in CKD with oxidative stress and autonomic neuron hyperactivity exhibited calcium handling abnormalities, which contributed to the induction of VT.
Collapse
Affiliation(s)
- Shih-Yu Huang
- Division of Cardiac Electrophysiology, Cardiovascular Center, Cathay General Hospital, Taipei City, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei City, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei City, Taiwan.,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
| | - Yen-Yu Lu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.,Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Urasoe, Japan
| | - Shih-Ann Chen
- Division of Cardiology, Department of Medicine, Heart Rhythm Center, Taipei Veterans General Hospital, Taipei City, Taiwan.,Cardiovascular Center, Taichung Veterans General Hospital, Taichung City, 40705, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei City, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan.,Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan.,Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
| |
Collapse
|
12
|
Ventricular arrhythmias in mouse models of diabetic kidney disease. Sci Rep 2021; 11:20570. [PMID: 34663875 PMCID: PMC8523538 DOI: 10.1038/s41598-021-99891-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 12/05/2022] Open
Abstract
Chronic kidney disease (CKD) affects more than 20 million people in the US, and it is associated with a significantly increased risk of sudden cardiac death (SCD). Despite the significance, the mechanistic relationship between SCD and CKD is not clear and there are few effective therapies. Using optical mapping techniques, we tested the hypothesis that mouse models of progressive diabetic kidney disease (DKD) exhibit enhanced ventricular arrhythmia incidence and underlying arrhythmia substrates. Compared to wild-type mice, both Leprdb/db eNOS−/− (2KO) and high fat diet plus low dose streptozotocin (HFD + STZ) mouse models of DKD experienced sudden death and greater arrhythmia inducibility, which was more common with isoproterenol than programmed electrical stimulation. 2KO mice demonstrated slowed conduction velocity, prolonged action potential duration (APD), and myocardial fibrosis; both 2KO and HFD + STZ mice exhibited arrhythmias and calcium dysregulation with isoproterenol challenge. Finally, circulating concentrations of the uremic toxin asymmetric dimethylarginine (ADMA) were elevated in 2KO mice. Incubation of human cardiac myocytes with ADMA prolonged APD, as also observed in 2KO mice hearts ex vivo. The present study elucidates an arrhythmia-associated mechanism of sudden death associated with DKD, which may lead to more effective treatments in the vulnerable DKD patient population.
Collapse
|
13
|
Graves JM, Vallejo JA, Hamill CS, Wang D, Ahuja R, Patel S, Faul C, Wacker MJ. Fibroblast growth factor 23 (FGF23) induces ventricular arrhythmias and prolongs QTc interval in mice in an FGF receptor 4-dependent manner. Am J Physiol Heart Circ Physiol 2021; 320:H2283-H2294. [PMID: 33929896 DOI: 10.1152/ajpheart.00798.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is a phosphate regulating protein hormone released by osteocytes. FGF23 becomes markedly elevated in chronic kidney disease (CKD), for which the leading cause of death is cardiovascular disease, particularly sudden cardiac death. Previously, we found that FGF23 increases intracellular Ca2+ in cardiomyocytes and alters contractility in mouse ventricles ex vivo via FGF receptor 4 (FGFR4). In the present study, we demonstrate that FGF23 induces cardiac arrhythmias and prolongs QTc interval in mice, and we tested whether these effects are mediated through FGFR4. In isolated Langendorff perfused hearts, FGF23 perfusion increased mechanical arrhythmias in the form of premature ventricular beats (PVBs), and induced runs of ventricular tachycardia in 6 of 11 animals, which were attenuated with pretreatment of an anti-FGFR4 blocking antibody. Ex vivo ECG analysis of isolated intact hearts showed increased ventricular arrhythmias and QTc prolongation after FGF23 infusion compared with vehicle. In vivo, injection of FGF23 into the jugular vein led to the emergence of premature ventricular contractions (PVCs) in 5 out of 11 experiments. FGF23 also produced a significant lengthening effect upon QTc interval in vivo. In vivo FGFR4 blockade ameliorated the arrhythmogenic and QTc prolonging effects of FGF23. Finally, FGF23 increased cardiomyocyte Ca2+ levels in intact left ventricular muscle which was inhibited by FGR4 blockade. We conclude that FGF23/FGFR4 signaling in the heart may contribute to ventricular arrhythmogenesis and repolarization disturbances commonly observed in patients with CKD via Ca2+ overload and may be an important therapeutic target to reduce cardiac mortality in CKD.NEW & NOTEWORTHY Here we provide direct evidence that fibroblast growth factor 23 (FGF23), a phosphaturic hormone elevated in chronic kidney disease, is proarrhythmic. FGF23 acutely triggered ventricular arrhythmias and prolonged corrected QT interval (QTc) in isolated mouse hearts and in vivo. FGF23 also increased Ca2+ levels in ventricular muscle tissue. Blockade of the FGF receptor 4 signaling pathway using a monoclonal antibody ameliorated ventricular arrhythmias, QTc prolongation, and elevated ventricular Ca2+ induced by FGF23, and may represent a potential therapeutic target in chronic kidney disease.
Collapse
Affiliation(s)
- Jonah M Graves
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Julian A Vallejo
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Chelsea S Hamill
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Derek Wang
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Rohan Ahuja
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Shaan Patel
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Christian Faul
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael J Wacker
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| |
Collapse
|
14
|
Weighted gene co-expression network analysis of chronic kidney disease and hemodialysis patients. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
15
|
Law JP, Price AM, Pickup L, Radhakrishnan A, Weston C, Jones AM, McGettrick HM, Chua W, Steeds RP, Fabritz L, Kirchhof P, Pavlovic D, Townend JN, Ferro CJ. Clinical Potential of Targeting Fibroblast Growth Factor-23 and αKlotho in the Treatment of Uremic Cardiomyopathy. J Am Heart Assoc 2020; 9:e016041. [PMID: 32212912 PMCID: PMC7428638 DOI: 10.1161/jaha.120.016041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease is highly prevalent, affecting 10% to 15% of the adult population worldwide and is associated with increased cardiovascular morbidity and mortality. As chronic kidney disease worsens, a unique cardiovascular phenotype develops characterized by heart muscle disease, increased arterial stiffness, atherosclerosis, and hypertension. Cardiovascular risk is multifaceted, but most cardiovascular deaths in patients with advanced chronic kidney disease are caused by heart failure and sudden cardiac death. While the exact drivers of these deaths are unknown, they are believed to be caused by uremic cardiomyopathy: a specific pattern of myocardial hypertrophy, fibrosis, with both diastolic and systolic dysfunction. Although the pathogenesis of uremic cardiomyopathy is likely to be multifactorial, accumulating evidence suggests increased production of fibroblast growth factor-23 and αKlotho deficiency as potential major drivers of cardiac remodeling in patients with uremic cardiomyopathy. In this article we review the increasing understanding of the physiology and clinical aspects of uremic cardiomyopathy and the rapidly increasing knowledge of the biology of both fibroblast growth factor-23 and αKlotho. Finally, we discuss how dissection of these pathological processes is aiding the development of therapeutic options, including small molecules and antibodies, directly aimed at improving the cardiovascular outcomes of patients with chronic kidney disease and end-stage renal disease.
Collapse
Affiliation(s)
- Jonathan P. Law
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Anna M. Price
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Luke Pickup
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Ashwin Radhakrishnan
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
| | - Chris Weston
- Institute of Immunology and ImmunotherapyUniversity of BirminghamUnited Kingdom
- NIHR Birmingham Biomedical Research CentreUniversity Hospitals Birmingham NHS Foundation Trust and University of BirminghamUnited Kingdom
| | - Alan M. Jones
- School of PharmacyUniversity of BirminghamUnited Kingdom
| | | | - Winnie Chua
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Richard P. Steeds
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Larissa Fabritz
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Paulus Kirchhof
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Davor Pavlovic
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Jonathan N. Townend
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Charles J. Ferro
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| |
Collapse
|
16
|
Ke HY, Chin LH, Tsai CS, Lin FZ, Chen YH, Chang YL, Huang SM, Chen YC, Lin CY. Cardiac calcium dysregulation in mice with chronic kidney disease. J Cell Mol Med 2020; 24:3669-3677. [PMID: 32064746 PMCID: PMC7131917 DOI: 10.1111/jcmm.15066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular complications are leading causes of morbidity and mortality in patients with chronic kidney disease (CKD). CKD significantly affects cardiac calcium (Ca2+) regulation, but the underlying mechanisms are not clear. The present study investigated the modulation of Ca2+ homeostasis in CKD mice. Echocardiography revealed impaired fractional shortening (FS) and stroke volume (SV) in CKD mice. Electrocardiography showed that CKD mice exhibited longer QT interval, corrected QT (QTc) prolongation, faster spontaneous activities, shorter action potential duration (APD) and increased ventricle arrhythmogenesis, and ranolazine (10 µmol/L) blocked these effects. Conventional microelectrodes and the Fluo‐3 fluorometric ratio techniques indicated that CKD ventricular cardiomyocytes exhibited higher Ca2+ decay time, Ca2+ sparks, and Ca2+ leakage but lower [Ca2+]i transients and sarcoplasmic reticulum Ca2+ contents. The CaMKII inhibitor KN93 and ranolazine (RAN; late sodium current inhibitor) reversed the deterioration in Ca2+ handling. Western blots revealed that CKD ventricles exhibited higher phosphorylated RyR2 and CaMKII and reduced phosphorylated SERCA2 and SERCA2 and the ratio of PLB‐Thr17 to PLB. In conclusions, the modulation of CaMKII, PLB and late Na+ current in CKD significantly altered cardiac Ca2+ regulation and electrophysiological characteristics. These findings may apply on future clinical therapies.
Collapse
Affiliation(s)
- Hung-Yen Ke
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Li-Han Chin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Zhi Lin
- Grade institute of life sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering and Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
17
|
Chan YH, Chang GJ, Lai YJ, Chen WJ, Chang SH, Hung LM, Kuo CT, Yeh YH. Atrial fibrillation and its arrhythmogenesis associated with insulin resistance. Cardiovasc Diabetol 2019; 18:125. [PMID: 31558158 PMCID: PMC6761716 DOI: 10.1186/s12933-019-0928-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/14/2019] [Indexed: 02/07/2023] Open
Abstract
Background Insulin resistance (IR) is considered as a risk factor for atrial fibrillation (AF) even before diabetes develops. The pathophysiology and underlying mechanism are largely unclear. Methods We investigated the corresponding mechanism in two IR models of rats fed 15-week high-fat (HFa) and high-fructose/cholesterol (HFr) diets. AF was evaluated and induced by burst atrial pacing. Isolated atrial myocytes were used for whole-cell patch clamp and calcium assessment. Ex vivo whole heart was used for optical mapping. Western blot and immunofluorescence were used for quantitative protein evaluation. Results Both HFa and HFr rat atria were vulnerable to AF evaluated by burst atrial pacing. Isolated atrial myocytes from HFa and HFr rats revealed significantly increased sarcoplasmic reticulum calcium content and diastolic calcium sparks. Whole-heart mapping showed prolonged calcium transient duration, conduction velocity reduction, and repetitive ectopic focal discharge in HFa and HFr atria. Protein analysis revealed increased TGF-β1 and collagen expression; increased superoxide production; abnormal upregulation of calcium-homeostasis-related proteins, including oxidized CaMKIIδ, phosphorylated-phospholamban, phosphorylated-RyR-2, and sodium-calcium exchanger; and increased Rac1 activity in both HFa and HFr atria. We observed that inhibition of CaMKII suppressed AF in both HF and HFr diet-fed rats. In vitro palmitate-induced IR neonatal cardiomyocytes and atrial fibroblasts expressed significantly more TGF-β1 than did controls, suggesting paracrine and autocrine effects on both myocytes and fibroblasts. Conclusions IR engenders both atrial structural remodeling and abnormal intracellular calcium homeostasis, contributing to increased AF susceptibility. The inhibition of CaMKII may be a potential therapeutic target for AF in insulin resistance.
Collapse
Affiliation(s)
- Yi-Hsin Chan
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Microscopy Core Laboratory, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University, Taoyuan, Taiwan
| | - Ying-Ju Lai
- Department of Respiratory Therapy, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Center for Big Data Analytics and Statistics, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Man Hung
- Department of Biomedical Sciences, College of Medicine, Healthy and Aging Research Center, Chang-Gung University, Taoyuan, Taiwan
| | - Chi-Tai Kuo
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. .,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. .,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.
| |
Collapse
|
18
|
Zhao SQ, Shen ZC, Gao BF, Han P. microRNA-206 overexpression inhibits epithelial-mesenchymal transition and glomerulosclerosis in rats with chronic kidney disease by inhibiting JAK/STAT signaling pathway. J Cell Biochem 2019; 120:14604-14617. [PMID: 31148248 DOI: 10.1002/jcb.28722] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/06/2019] [Accepted: 03/15/2019] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is a traumatic disease with significant psychic consequences to the patient's overall physical condition. microRNA-206 (miR-206) has been reported to play an essential role in the development of various diseases. The purpose of the present study is to investigate the effect of miR-206 through the JAK/STAT signaling pathway on epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells and glomerulosclerosis in rats with CKD. The targeting relationship between miR-206 and ANXA1 was verified. To explore the role of miR-206 in CKD, the model of CKD rats was established to detect glomerular sclerosis index (GSI), contents of interleukin-6 (IL-6) and transforming growth factor-beta1 (TGF-β1), and expression of type IV collagen. Moreover, to further determine the roles of both miR-206 and the JAK/STAT signaling pathway in CKD, the gain- and loss-of function approaches were performed with the expression of ANXA1, α-SMA, E-cadherin, vimentin, N-cadherin, and the JAK/STAT signaling pathway-related genes detected. miR-206 negatively targeted ANXA1. Overexpressed miR-206 inhibited the degeneration and interstitial fibrosis of renal tubular epithelial cells, decreased GSI of rats, and the expression of type IV collagen, TGF-β1 and IL-6. Overexpressed miR-206 inhibited the degeneration of renal tubular epithelial cells, the expression of ANXA1, α-SMA, TGF-β1, p-STAT3, STAT3, p-STAT1, STAT1, p-JAK2, and JAK2, while promoted the expression of E-cadherin. Taken together the results, miR-206 inhibits EMT of renal tubular epithelial cells and glomerulosclerosis by inactivating the JAK/STAT signaling pathway via ANXA1 in CKD.
Collapse
Affiliation(s)
- Shi-Qi Zhao
- Department of Emergency, Linyi People's Hospital, Linyi, China
| | - Zhao-Chun Shen
- Department of Emergency, The Third People's Hospital of Linyi, Linyi, China
| | - Bing-Feng Gao
- Department of Cardiology, The Third People's Hospital of Linyi, Linyi, China
| | - Ping Han
- Department of Respiratory Medicine, Linyi People's Hospital, Linyi, China
| |
Collapse
|
19
|
Verkaik M, Oranje M, Abdurrachim D, Goebel M, Gam Z, Prompers JJ, Helmes M, Ter Wee PM, van der Velden J, Kuster DW, Vervloet MG, Eringa EC. High Fibroblast Growth Factor 23 concentrations in experimental renal failure impair calcium handling in cardiomyocytes. Physiol Rep 2019; 6:e13591. [PMID: 29611320 PMCID: PMC5880876 DOI: 10.14814/phy2.13591] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
The overwhelming majority of patients with chronic kidney disease (CKD) die prematurely before reaching end‐stage renal disease, mainly due to cardiovascular causes, of which heart failure is the predominant clinical presentation. We hypothesized that CKD‐induced increases of plasma FGF23 impair cardiac diastolic and systolic function. To test this, mice were subjected to 5/6 nephrectomy (5/6Nx) or were injected with FGF23 for seven consecutive days. Six weeks after surgery, plasma FGF23 was higher in 5/6Nx mice compared to sham mice (720 ± 31 vs. 256 ± 3 pg/mL, respectively, P = 0.034). In cardiomyocytes isolated from both 5/6Nx and FGF23 injected animals the rise of cytosolic calcium during systole was slowed (−13% and −19%, respectively) as was the decay of cytosolic calcium during diastole (−15% and −21%, respectively) compared to controls. Furthermore, both groups had similarly decreased peak cytosolic calcium content during systole. Despite lower cytosolic calcium contents in CKD or FGF23 pretreated animals, no changes were observed in contractile parameters of cardiomyocytes between the groups. Expression of calcium handling proteins and cardiac troponin I phosphorylation were similar between groups. Blood pressure, the heart weight:tibia length ratio, α‐MHC/β‐MHC ratio and ANF mRNA expression, and systolic and diastolic function as measured by MRI did not differ between groups. In conclusion, the rapid, CKD‐induced rise in plasma FGF23 and the similar decrease in cardiomyocyte calcium transients in modeled kidney disease and following 1‐week treatment with FGF23 indicate that FGF23 partly mediates cardiomyocyte dysfunction in CKD.
Collapse
Affiliation(s)
- Melissa Verkaik
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Maarten Oranje
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Max Goebel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Zeineb Gam
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michiel Helmes
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Pieter M Ter Wee
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Diederik W Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc G Vervloet
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
20
|
Poulikakos D, Hnatkova K, Skampardoni S, Green D, Kalra P, Malik M. Sudden Cardiac Death in Dialysis: Arrhythmic Mechanisms and the Value of Non-invasive Electrophysiology. Front Physiol 2019; 10:144. [PMID: 30873044 PMCID: PMC6401645 DOI: 10.3389/fphys.2019.00144] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 02/07/2019] [Indexed: 01/10/2023] Open
Abstract
Sudden Cardiac Death (SCD) is the leading cause of cardiovascular death in dialysis patients. This review discusses potential underlying arrhythmic mechanisms of SCD in the dialysis population. It examines recent evidence from studies using implantable loop recorders and from electrophysiological studies in experimental animal models of chronic kidney disease. The review summarizes advances in the field of non-invasive electrophysiology for risk prediction in dialysis patients focusing on the predictive value of the QRS-T angle and of the assessments of autonomic imbalance by means of heart rate variability analysis. Future research directions in non-invasive electrophysiology are identified to advance the understanding of the arrhythmic mechanisms. A suggestion is made of incorporation of non-invasive electrophysiology procedures into clinical practice. Key Concepts: - Large prospective studies in dialysis patients with continuous ECG monitoring are required to clarify the underlying arrhythmic mechanisms of SCD in dialysis patients. - Obstructive sleep apnoea may be associated with brady-arrhythmias in dialysis patients. Studies are needed to elucidate the burden and impact of sleeping disorders on arrhythmic complications in dialysis patients. - The QRS-T angle has the potential to be used as a descriptor of uremic cardiomyopathy. - The QRS-T angle can be calculated from routine collected surface ECGs. Multicenter collaboration is required to establish best methodological approach and normal values. - Heart Rate Variability provides indirect assessment of cardiac modulation that may be relevant for cardiac risk prediction in dialysis patients. Short-term recordings with autonomic provocations are likely to overcome the limitations of out of hospital 24-h recordings and should be prospectively assessed.
Collapse
Affiliation(s)
- Dimitrios Poulikakos
- Renal Department, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Centre for Cardiac Research, Institute of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| | - Katerina Hnatkova
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sofia Skampardoni
- Renal Department, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Centre for Cardiac Research, Institute of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| | - Darren Green
- Renal Department, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Centre for Cardiac Research, Institute of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| | - Philip Kalra
- Renal Department, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Centre for Cardiac Research, Institute of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| | - Marek Malik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Skampardoni S, Green D, Hnatkova K, Malik M, Kalra PA, Poulikakos D. QRS-T Angle Predicts Cardiac Risk and Correlates With Global Longitudinal Strain in Prevalent Hemodialysis Patients. Front Physiol 2019; 10:145. [PMID: 30858805 PMCID: PMC6397862 DOI: 10.3389/fphys.2019.00145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 02/07/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Cardiovascular disease is the commonest cause of death in hemodialysis (HD) patients but accurate risk prediction is lacking. The spatial QRS – T angle is a promising electrophysiological marker for sudden cardiac death risk stratification. The aim of this study was to assess the prognostic value of spatial QRS-T angle derived from standard 12 lead electrocardiograms (ECG) and its association with echocardiographic parameters in HD patients. Methods: This prospective study of 178 prevalent HD patients (aged 67 ± 14 years, 72% men) collected ECG and echocardiographic data on an annual basis. Baseline echocardiograms at study entry were used for cross-sectional comparisons with ECGs. Study endpoints were all-cause mortality, cardiovascular mortality, and major adverse cardiac events (MACE). The QRS – T angle was calculated from standard 10-s ECG as the total cosine R to T (TCRT) using singular value decomposition and expressed in degrees. TCRT above 100° was defined as abnormal. Results: During a follow-up period of 36 ± 19 months, 74 patients died, including 17 cardiac deaths, and 54 suffered from MACE. In multivariate Cox regression analysis, QRS-T angle by TCRT at baseline was associated with increased cardiovascular mortality both as a continuous value and dichotomized below or above 100° (HR 1.016, p = 0.029, CI: 1.002–1.030 and HR 3.506, CI: 1.118–10.995, p = 0.031 respectively) and with MACE dichotomized at 100° (HR 1.902, CI: 1.046–3.459; p = 0.035). In multivariate regression analysis including baseline parameters, echocardiographic global longitudinal strain (GLS) was significantly correlated with TCRT (F 9.648, r2 = 0.192, standardized β = 0.331, unstandardized β = 3.567, t = 4.4429, CI: 1.976–5.157, p < 0.001). Conclusion: TCRT correlates with GLS and is independently associated with cardiac deaths and MACE in HD patients.
Collapse
Affiliation(s)
- Sofia Skampardoni
- Department of Renal Medicine, Salford Royal NHS Foundation Trust and The University of Manchester, Manchester, United Kingdom
| | - Darren Green
- Department of Renal Medicine, Salford Royal NHS Foundation Trust and The University of Manchester, Manchester, United Kingdom
| | - Katerina Hnatkova
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Marek Malik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Philip A Kalra
- Department of Renal Medicine, Salford Royal NHS Foundation Trust and The University of Manchester, Manchester, United Kingdom
| | - Dimitrios Poulikakos
- Department of Renal Medicine, Salford Royal NHS Foundation Trust and The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
22
|
Moe SM. Sudden cardiac death in patients undergoing dialysis: More than a single toxin. Heart Rhythm 2019; 16:318-319. [DOI: 10.1016/j.hrthm.2018.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Indexed: 10/28/2022]
|
23
|
Wu CI, Chang SL, Lin CY, Vicera JJB, Lin YJ, Lo LW, Chung FP, Hu YF, Chang TY, Chao TF, Liao JN, Tuan TC, Liu CM, Te ALD, Chen SA. Clinical significance of J wave in prediction of ventricular arrhythmia in patients with acute myocardial infarction. J Cardiol 2019; 73:351-357. [PMID: 30595403 DOI: 10.1016/j.jjcc.2018.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/14/2018] [Accepted: 11/26/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND J wave syndrome and myocardial ischemia are related with malignant ventricular arrhythmia (VA). The characteristics of dynamic J wave in patients with early phase of acute myocardial infarction (AMI) and subsequent VA or electrical storm (ES) have not been well evaluated. OBJECTIVE We investigated the utility of J wave in the prediction of VA and ES in patients within the early phase of AMI. METHODS This study retrospectively enrolled 208 patients (mean age 69±15 years, 171 males) with AMI. Of them, 50 patients had experienced VA during hospitalization and 24 had ES. The clinical and electrocardiographic characteristics of these patients with and without VA were compared. RESULTS Patients with VA had a higher incidence of chronic kidney disease (CKD) and J wave compared with those without VA. The hazard ratio (HR) of J wave for VA was 4.31 (p<0.01) and CKD was 2.64 (p<0.01). In the VA group, ES patients had a higher incidence of diabetes mellitus (DM) (HR 2.73, p=0.02) and J wave (HR 4.21, p<0.01). If the AMI patients had J wave, the OR for mortality was 2.14 (p=0.03), VA events was 6.23 (p<0.01), and ES events was 12.15 (p<0.01). If VA patients had J wave, the mortality rate will significantly increase (OR 68.62, p=0.01). CONCLUSION The AMI patients who develop VA in the early phase of AMI had a higher incidence of J wave and CKD, and those who develop ES had a higher incidence of J wave and DM. It seems that J wave in AMI patients is a poor prognostic factor, and we found that J wave will increase mortality, VA events, and ES events. The majority locations of J wave were inferior leads although there was no relationship between the locations and VA incidence. If the VA patients had inferior or lateral J wave, it would further increase the risk of mortality.
Collapse
Affiliation(s)
- Cheng-I Wu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Shih-Lin Chang
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Chin-Yu Lin
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jennifer Jeanne B Vicera
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yenn-Jiang Lin
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Li-Wei Lo
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Fa-Po Chung
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Yu-Feng Hu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Ting-Yung Chang
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tze-Fan Chao
- Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Jo-Nan Liao
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Ta-Chuan Tuan
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chih-Min Liu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Abigail Louise D Te
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| |
Collapse
|
24
|
Thomsen MB, Nielsen MS, Aarup A, Bisgaard LS, Pedersen TX. Uremia increases QRS duration after β-adrenergic stimulation in mice. Physiol Rep 2018; 6:e13720. [PMID: 29984555 PMCID: PMC6036105 DOI: 10.14814/phy2.13720] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/28/2018] [Indexed: 01/14/2023] Open
Abstract
Chronic kidney disease (CKD) and uremia increase the risk of heart disease and sudden cardiac death. Coronary artery disease can only partly account for this. The remaining mechanistic links between CKD and sudden death are elusive, but may involve cardiac arrhythmias. For the present study, we hypothesized that a thorough electrophysiological study in mice with CKD would provide us valuable information that could aid in the identification of additional underlying causes of sudden cardiac death in patients with kidney disease. Partial (5/6) nephrectomy (NX) in mice induced mild CKD: plasma urea in NX was 24 ± 1 mmol/L (n = 23) versus 12 ± 1 mmol/L (n = 22) in sham-operated control mice (P < 0.05). Echocardiography did not identify structural or mechanical remodeling in NX mice. Baseline ECG parameters were comparable in conscious NX and control mice; however, the normal 24-h diurnal rhythm in QRS duration was lost in NX mice. Moreover, β-adrenergic stimulation (isoprenaline, 200 μg/kg intraperitoneally) prolonged QRS duration in conscious NX mice (from 12 ± 1 to 15 ± 2 msec, P < 0.05), but not in sham-operated controls (from 13 ± 1 to 13 ± 2 msec, P > 0.05). No spontaneous arrhythmias were observed in conscious NX mice, and intracardiac pacing in anesthetized mice showed a comparable arrhythmia vulnerability in NX and sham-operated mice. Isoprenaline (2 mg/kg intraperitoneally) changed the duration of the QRS complex from 11.2 ± 0.4 to 11.9 ± 0.5 (P = 0.06) in NX mice and from 10.7 ± 0.6 to 10.6 ± 0.6 (P = 0.50) in sham-operated mice. Ex vivo measurements of cardiac ventricular conduction velocity were comparable in NX and sham mice. Transcriptional activity of Scn5a, Gja1 and several profibrotic genes was similar in NX and sham mice. We conclude that proper kidney function is necessary to maintain diurnal variation in QRS duration and that sympathetic regulation of the QRS duration is altered in kidney disease.
Collapse
Affiliation(s)
- Morten B. Thomsen
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Morten S. Nielsen
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Annemarie Aarup
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Line S. Bisgaard
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Tanja X. Pedersen
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
25
|
Aref MW, Swallow EA, Chen NX, Moe SM, Allen MR. Skeletal vascular perfusion is altered in chronic kidney disease. Bone Rep 2018; 8:215-220. [PMID: 29955640 PMCID: PMC6020396 DOI: 10.1016/j.bonr.2018.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/04/2018] [Accepted: 05/02/2018] [Indexed: 11/25/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at an alarming risk of cardiovascular disease and fracture-associated mortality. CKD has been shown to have negative effects on vascular reactivity and organ perfusion. Although alterations in bone blood flow are linked to dysregulation of bone remodeling and mass in multiple conditions, changes to skeletal perfusion in the setting of CKD have not been explored. The goal of this study was to establish the effect of CKD on skeletal perfusion in a rat model of CKD. In two experiments with endpoints at 30 and 35 weeks of age, respectively, normal (NL) and Cy/+ (CKD) animals (n = 6/group) underwent in vivo intra-cardiac fluorescent microsphere injection to assess bone tissue perfusion. These two separate time points aimed to describe skeletal perfusion at 30 and 35 weeks based on previous studies demonstrating significant progression of hyperparthyroid bone disease during this timeframe. CKD animals had blood urea nitrogen (BUN) levels significantly higher than NL at both 30 and 35 weeks. At 30 weeks, perfusion was significantly higher in the femoral cortex (+259%, p < 0.05) but not in the tibial cortex (+140%, p = 0.11) of CKD animals relative to NL littermates. Isolated tibial marrow perfusion at 30 weeks showed a trend toward being higher (+183%, p = 0.08) in CKD. At 35 weeks, perfusion was significantly higher in both the femoral cortex (+173%, p < 0.05) and the tibial cortex (+241%, p < 0.05) in CKD animals when compared to their normal littermates. Isolated tibial marrow perfusion (-57%, p <0.05) and vertebral body perfusion (-71%, p <0.05) were lower in CKD animals. The current study demonstrates two novel findings regarding bone perfusion in an animal model of high turnover CKD. First, cortical bone perfusion in CKD animals is higher than in normal animals. Second, alterations in bone marrow perfision differed among the stages of CKD and were distinct from perfusion to the cortical bone. Determining whether these changes in bone perfusion are drivers, propagators, or consequences of skeletal deterioration in CKD will necessitate further work.
Collapse
Affiliation(s)
- Mohammad W. Aref
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Elizabeth A. Swallow
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Neal X. Chen
- Department of Medicine, – Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sharon M. Moe
- Department of Medicine, – Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States
| | - Matthew R. Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medicine, – Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biomedical Engineering, Indiana University Purdue University of Indianapolis, Indianapolis, IN, United States
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States
| |
Collapse
|
26
|
Abstract
Disordered calcium balance and homeostasis are common in patients with chronic kidney disease. Such alterations are commonly associated with abnormal bone remodeling, directly and indirectly. Similarly, positive calcium balance may also be a factor in the pathogenesis of extra skeletal soft tissue and arterial calcification. Calcium may directly affect cardiac structure and function through direct effects to alter cell signaling due to abnormal intracellular calcium homeostasis 2) extra-skeletal deposition of calcium and phosphate in the myocardium and small cardiac arterioles, 3) inducing cardiomyocyte hypertrophy through calcium and hormone activation of NFAT signaling mechanisms, and 4) increased aorta calcification resulting in chronic increased afterload leading to hypertrophy. Similarly, calcium may alter vascular smooth muscle cell function and affect cell signaling which may predispose to a proliferative phenotype important in arteriosclerosis and arterial calcification. Thus, disorders of calcium balance and homeostasis due to CKD-MBD may play a role in the high cardiovascular burden observed in patients with CKD.
Collapse
Affiliation(s)
- Sharon M Moe
- Indiana University School of Medicine, Division of Nephrology, Indianapolis, IN, United States; Department of Medicine, Roudebush Veterans Affairs Medical Center, Indianapolis, IN, United States.
| |
Collapse
|
27
|
Fu L, Zhou Q, Zhu W, Lin H, Ding Y, Shen Y, Hu J, Hong K. Do Implantable Cardioverter Defibrillators Reduce Mortality in Patients With Chronic Kidney Disease at All Stages? Int Heart J 2017; 58:371-377. [PMID: 28539571 DOI: 10.1536/ihj.16-357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
The benefits of implantable cardioverter defibrillator (ICD) implantation in chronic kidney disease (CKD) patients with high sudden cardiac death (SCD) risk are uncertain. To clarify the effects of receiving an ICD in CKD patients, we conducted this meta-analysis to identify the effects of ICDs on patients with CKD, including those on dialysis. We searched the Cochrane library, EMBASE, PubMed, and clinical trials for studies published before July 2016. Eleven studies including 20,196 CKD patients were considered for inclusion. The pooled analysis suggested that patients with an estimated glomerular filtration rate (eGFR) < 60 mL/minute/1.73 m2 would benefit from receiving treatments with ICDs compared with patients without an ICD device (aHR = 0.74; 95% confidence interval [CI], 0.63 to 0.86). [corrected]. This is the first report of a subgroup analysis on the survival rate of ICD implantation in CKD patients according to an eGFR group. The subgroup analysis indicated a similar protective association of ICDs in stage 3 (aHR = 0.71; 95% CI, 0.61 to 0.82) and 5 (aHR = 0.71; 95% CI, 0.54 to 0.92) CKD patients [corrected] compared with the control group. However, there was no significant improvement in all-cause mortality in stage 4 CKD patients (aHR = 1.02; 95%CI, 0.75 to 1.37) [corrected]. This is the first meta-analysis reporting that ICD implantation reduces all-cause mortality in stage 3 and 5 [corrected] CKD patients. However, the data do not indicate there is any benefit to ICD implantation in stage 4 [corrected] CKD patients.
Collapse
MESH Headings
- Death, Sudden, Cardiac/epidemiology
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/prevention & control
- Defibrillators, Implantable
- Global Health
- Humans
- Incidence
- Registries
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/diagnosis
- Renal Insufficiency, Chronic/mortality
- Risk Assessment
- Risk Factors
- Survival Rate/trends
Collapse
Affiliation(s)
- Linghua Fu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Qiongqiong Zhou
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Wengen Zhu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Huang Lin
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Ying Ding
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Yang Shen
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Jinzhu Hu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
| | - Kui Hong
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University
- Jiangxi Key Laboratory of Molecular Medicine
| |
Collapse
|
28
|
Jin H, Welzig CM, Aronovitz M, Noubary F, Blanton R, Wang B, Rajab M, Albano A, Link MS, Noujaim SF, Park HJ, Galper JB. QRS/T-wave and calcium alternans in a type I diabetic mouse model for spontaneous postmyocardial infarction ventricular tachycardia: A mechanism for the antiarrhythmic effect of statins. Heart Rhythm 2017; 14:1406-1416. [PMID: 28522367 DOI: 10.1016/j.hrthm.2017.05.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND The incidence of sudden arrhythmic death is markedly increased in diabetics. OBJECTIVE The purpose of this study was to develop a mouse model for postmyocardial infarction (post-MI) ventricular tachycardia (VT) in the diabetic heart and determine the mechanism of an antiarrhythmic effect of statins. METHODS ECG transmitters were implanted in wild-type (WT), placebo, and pravastatin-treated type I diabetic Akita mice. MIs were induced by coronary ligation, and Ca2+ transients were studied by optical mapping, and Ca2+ transients and sparks in left ventricular myocytes (VM) by the Ionoptix system and confocal microscopy. RESULTS Burst pacing of Akita mouse hearts resulted in rate-related QRS/T-wave alternans, which was attenuated in pravastatin-treated mice. Post-MI Akita mice developed QRS/T-wave alternans and VT at 2820 ± 879 beats per mouse, which decreased to 343 ± 115 in pravastatin-treated mice (n = 13, P <.05). Optical mapping demonstrated pacing-induced VT originating in the peri-infarction zone and Ca2+ alternans, both attenuated in hearts of statin-treated mice. Akita VM displayed Ca2+ alternans, and triggered activity as well as increased Ca2+ transient decay time (Tau), Ca2+ sparks, and cytosolic Ca2+ and decreased SR Ca2+ stores all of which were in part reversed in cells from statin treated mice. Homogenates of Akita ventricles demonstrated decreased SERCA2a/PLB ratio and increased ratio of protein phosphatase (PP-1) to the PP-1 inhibitor PPI-1 which were reversed in homogenates of pravastatin-treated Akita mice. CONCLUSION Pravastatin decreased the incidence of post-MI VT and Ca2+ alternans in Akita mouse hearts in part by revering abnormalities of Ca2+ handling via the PP-1/PPI-1 pathway.
Collapse
Affiliation(s)
- Hongwei Jin
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts.
| | - Charles M Welzig
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Departments of Neurology, Physiology and Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mark Aronovitz
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Farzad Noubary
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Tufts Clinical and Translational Science Institute, Boston, Massachusetts
| | - Robert Blanton
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts; Cardiovascular Division, Cardiovascular Center, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Bo Wang
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Mohammad Rajab
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts; Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Alfred Albano
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts; Spectrum Health, Grand Rapids, Michigan
| | - Mark S Link
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Tufts Clinical and Translational Science Institute, Boston, Massachusetts; UT Southwestern Medical Center, Dallas, Texas
| | - Sami F Noujaim
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, Tampa, Florida
| | - Ho-Jin Park
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts.
| | - Jonas B Galper
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts; Cardiovascular Division, Cardiovascular Center, Department of Medicine, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
29
|
Electronegative LDL-mediated cardiac electrical remodeling in a rat model of chronic kidney disease. Sci Rep 2017; 7:40676. [PMID: 28094801 PMCID: PMC5240592 DOI: 10.1038/srep40676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/09/2016] [Indexed: 12/11/2022] Open
Abstract
The mechanisms underlying chronic kidney disease (CKD)–associated higher risks for life-threatening ventricular tachyarrhythmias remain poorly understood. In rats subjected to unilateral nephrectomy (UNx), we examined cardiac electrophysiological remodeling and relevant mechanisms predisposing to ventricular arrhythmias. Adult male Sprague-Dawley rats underwent UNx (n = 6) or sham (n = 6) operations. Eight weeks later, the UNx group had higher serum blood urea nitrogen and creatinine levels and a longer electrocardiographic QTc interval than did the sham group. Patch-clamp studies revealed epicardial (EPI)-predominant prolongation of the action potential duration (APD) at 50% and 90% repolarization in UNx EPI cardiomyocytes compared to sham EPI cardiomyocytes. A significant reduction of the transient outward potassium current (Ito) in EPI but not in endocardial (ENDO) cardiomyocytes of UNx rats led to a decreased transmural gradient of Ito. The reduction of Ito currents in UNx EPI cardiomyocytes was secondary to downregulation of KChIP2 but not Kv4.2, Kv4.3, and Kv1.4 protein expression. Incubation of plasma electronegative low-density lipoprotein (LDL) from UNx rats with normal EPI and ENDO cardiomyocytes recapitulated the electrophysiological phenotype of UNx rats. In conclusion, CKD disrupts the physiological transmural gradient of Ito via downregulation of KChIP2 proteins in the EPI region, which may promote susceptibility to ventricular tachyarrhythmias. Electronegative LDL may underlie downregulation of KChIP2 in CKD.
Collapse
|
30
|
Lu JL, Molnar MZ, Ma JZ, George LK, Sumida K, Kalantar-Zadeh K, Kovesdy CP. Racial Differences in Association of Serum Calcium with Mortality and Incident Cardio- and Cerebrovascular Events. J Clin Endocrinol Metab 2016; 101:4851-4859. [PMID: 27631543 PMCID: PMC5155693 DOI: 10.1210/jc.2016-1802] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Abnormalities in calcium metabolism may potentially contribute to the development of vascular disease. Calcium metabolism may be different in African American (AA) vs white individuals, but the effect of race on the association of serum calcium with clinical outcomes remains unclear. OBJECTIVE This study sought to examine race-specific associations of serum calcium levels with mortality and with major incident cardiovascular events. DESIGN AND SETTING This was a historical cohort study in the U.S. Department of Veterans Affairs health care facilities. PARTICIPANTS Participants included veterans (n = 1 967 622) with estimated glomerular filtration rate ≥ 60 mL/min/1.73 m2. MAIN OUTCOME MEASURES The association between serum calcium levels with all-cause mortality, incident coronary heart disease (CHD), and ischemic stroke incidence was examined in multivariable adjusted Cox proportional hazards models, including an interaction term for calcium and race. RESULTS The association of calcium with all-cause mortality was U-shaped in both AA and white patients, but race modified the association of calcium with all-cause mortality. Compared with white patients, AA patients experienced lower risk of mortality when calcium was ≥ 8.8 mg/dL, with a statistically significant interaction (P < .001). Conversely, AA vs white race was associated with higher mortality when calcium was < 8.8 mg/dL. Calcium showed no significant association with ischemic stroke or CHD in both races; and race did not modify these associations (P = .37 and 0.11, respectively for interaction term). CONCLUSIONS Race modified the U-shaped association between calcium and all-cause mortality. Serum calcium is not associated with incident stroke or CHD in either AA or white patients. The race-specific difference in the association of calcium levels with mortality warrants further examination.
Collapse
Affiliation(s)
- Jun Ling Lu
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| | - Miklos Z Molnar
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| | - Jennie Z Ma
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| | - Lekha K George
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| | - Keiichi Sumida
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| | - Csaba P Kovesdy
- Division of Nephrology (J.L.L., M.Z.M., L.K.G., K.S., C.P.K.), University of Tennessee Health Science Center, Memphis Tennessee 38163; Department of Public Health Sciences and Division of Nephrology, Department of Medicine (J.Z.M.), University of Virginia, Charlottesville, Virginia 22908; Nephrology Center (K.S.), Toranomon Hospital Kajigaya, Kanagawa 213-8587, Japan; Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension (K.K.-Z.), University of California-Irvine Medical Center, Orange, California 92868; and Nephrology Section (C.P.K.), Memphis VA Medical Center, Memphis, Tennessee 38104
| |
Collapse
|
31
|
Rutherford E, Talle MA, Mangion K, Bell E, Rauhalammi SM, Roditi G, McComb C, Radjenovic A, Welsh P, Woodward R, Struthers AD, Jardine AG, Patel RK, Berry C, Mark PB. Defining myocardial tissue abnormalities in end-stage renal failure with cardiac magnetic resonance imaging using native T1 mapping. Kidney Int 2016; 90:845-52. [PMID: 27503805 PMCID: PMC5035134 DOI: 10.1016/j.kint.2016.06.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/25/2016] [Accepted: 06/02/2016] [Indexed: 01/24/2023]
Abstract
Noninvasive quantification of myocardial fibrosis in end-stage renal disease is challenging. Gadolinium contrast agents previously used for cardiac magnetic resonance imaging (MRI) are contraindicated because of an association with nephrogenic systemic fibrosis. In other populations, increased myocardial native T1 times on cardiac MRI have been shown to be a surrogate marker of myocardial fibrosis. We applied this method to 33 incident hemodialysis patients and 28 age- and sex-matched healthy volunteers who underwent MRI at 3.0T. Native T1 relaxation times and feature tracking–derived global longitudinal strain as potential markers of fibrosis were compared and associated with cardiac biomarkers. Left ventricular mass indices were higher in the hemodialysis than the control group. Global, Septal and midseptal T1 times were all significantly higher in the hemodialysis group (global T1 hemodialysis 1171 ± 27 ms vs. 1154 ± 32 ms; septal T1 hemodialysis 1184 ± 29 ms vs. 1163 ± 30 ms; and midseptal T1 hemodialysis 1184 ± 34 ms vs. 1161 ± 29 ms). In the hemodialysis group, T1 times correlated with left ventricular mass indices. Septal T1 times correlated with troponin and electrocardiogram-corrected QT interval. The peak global longitudinal strain was significantly reduced in the hemodialysis group (hemodialysis -17.7±5.3% vs. -21.8±6.2%). For hemodialysis patients, the peak global longitudinal strain significantly correlated with left ventricular mass indices (R = 0.426), and a trend was seen for correlation with galectin-3, a biomarker of cardiac fibrosis. Thus, cardiac tissue properties of hemodialysis patients consistent with myocardial fibrosis can be determined noninvasively and associated with multiple structural and functional abnormalities.
Collapse
Affiliation(s)
- Elaine Rutherford
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK; University of Dundee, Division of Cardiovascular & Diabetes Medicine, Dundee, Scotland, UK.
| | - Mohammed A Talle
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Kenneth Mangion
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Elizabeth Bell
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Samuli M Rauhalammi
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Giles Roditi
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Christie McComb
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Aleksandra Radjenovic
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Rosemary Woodward
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Allan D Struthers
- University of Dundee, Division of Cardiovascular & Diabetes Medicine, Dundee, Scotland, UK
| | - Alan G Jardine
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Rajan K Patel
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Colin Berry
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| |
Collapse
|
32
|
Subcutaneous nerve activity and mechanisms of sudden death in a rat model of chronic kidney disease. Heart Rhythm 2015; 13:1105-1112. [PMID: 26744093 DOI: 10.1016/j.hrthm.2015.12.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND The mechanisms of sudden death in chronic kidney disease (CKD) remain unclear. OBJECTIVE The purpose of this study was to test the hypotheses that subcutaneous nerve activity (SCNA) can be used to estimate sympathetic tone in ambulatory rats and that abrupt reduction of SCNA precedes the spontaneous arrhythmic death of Cy/+ rats. METHODS Radiotransmitters were implanted in ambulatory normal (N = 6) and Cy/+ (CKD; N = 6) rats to record electrocardiogram and SCNA. Two additional rats were studied before and after chemical sympathectomy with 6-hydroxydopamine. RESULTS In normal rats, the baseline heart rate (HR) and SCNA were 351 ± 29 bpm and 5.12 ± 2.97 mV·s, respectively. SCNA abruptly increased HR by 4.31% (95% confidence interval 4.15%-4.47%). In comparison, the CKD rats had reduced baseline HR (336 ± 21 bpm, P < .01) and SCNA (4.27 ± 3.19 mV·s, P < .01). When SCNA was observed, HR increased by only 2.48% (confidence interval 2.29%-2.67%, P < .01). All Cy/+ rats died suddenly, preceded by sinus bradycardia, advanced (second- and third-degree) AV block (N = 6), and/or ventricular tachycardia or fibrillation (N = 3). Sudden death was preceded by a further reduction of SCNA (3.22 ± 2.86 mV·s, P < .01) and sinus bradycardia (243 ± 55 bpm, P < .01). Histologic studies in CKD rats showed myocardial calcification that involved the conduction system. Chemical sympathectomy resulted in progressive reduction of SCNA over 7 days. CONCLUSION SCNA can be used to estimate sympathetic tone in ambulatory rats. CKD is associated with reduced HR response to SCNA and conduction system diseases. Abrupt reduction of sympathetic tone precedes AV block, ventricular arrhythmia, and sudden death of CKD rats.
Collapse
|
33
|
Organ JM, Srisuwananukorn A, Price P, Joll JE, Biro KC, Rupert JE, Chen NX, Avin KG, Moe SM, Allen MR. Reduced skeletal muscle function is associated with decreased fiber cross-sectional area in the Cy/+ rat model of progressive kidney disease. Nephrol Dial Transplant 2015; 31:223-30. [PMID: 26442903 DOI: 10.1093/ndt/gfv352] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 09/08/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The combination of skeletal muscle wasting and compromised function plays a role in the health decline commonly observed in chronic kidney disease (CKD) patients, but the pathophysiology of muscle mass/strength changes remains unclear. The purpose of this study was to characterize muscle properties in the Cy/+ rat model of spontaneously progressive CKD. METHODS Leg muscle function and serum biochemistry of male Cy/+ (CKD) rats and their nonaffected littermates (NLs) were assessed in vivo at 25, 30 and 35 weeks of age. Architecture and histology of extensor digitorum longus (EDL) and soleus (SOL) muscles were assessed ex vivo at the conclusion of the experiment. We tested the hypothesis that animals with CKD have progressive loss of muscle function, and that this functional deficit is associated with loss of muscle mass and quality. RESULTS Thirty-five-week-old CKD rats produced significantly lower maximum torque in ankle dorsiflexion and shorter time to maximum torque, and longer half relaxation time in dorsiflexion and plantarflexion compared with NL rats. Peak dorsiflexion torque (but not plantarflexion torque) in CKD remained steady from 25 to 35 weeks, while in NL rats, peak torque increased. Mass, physiologic cross-sectional area (CSA) and fiber-type (myosin heavy chain isoform) proportions of EDL and SOL were not different between CKD and NL. However, the EDL of CKD rats showed reduced CSAs in all fiber types, while only MyHC-1 fibers were decreased in area in the SOL. CONCLUSIONS The results of this study demonstrate that muscle function progressively declines in the Cy/+ rat model of CKD. Because whole muscle mass and architecture do not vary between CKD and NL, but CKD muscles show reduction in individual fiber CSA, our data suggest that the functional decline is related to increased muscle fiber atrophy.
Collapse
Affiliation(s)
- Jason M Organ
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew Srisuwananukorn
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paige Price
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffery E Joll
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kelly C Biro
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joseph E Rupert
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Neal X Chen
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith G Avin
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Science, Indianapolis, IN, USA
| | - Sharon M Moe
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
34
|
Fontes MSC, Papazova DA, van Koppen A, de Jong S, Korte SM, Bongartz LG, Nguyen TQ, Bierhuizen MFA, de Boer TP, van Veen TAB, Verhaar MC, Joles JA, van Rijen HVM. Arrhythmogenic Remodeling in Murine Models of Deoxycorticosterone Acetate-Salt-Induced and 5/6-Subtotal Nephrectomy-Salt-Induced Cardiorenal Disease. Cardiorenal Med 2015. [PMID: 26195973 DOI: 10.1159/000430475] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Renal failure is associated with adverse cardiac remodeling and sudden cardiac death. The mechanism leading to enhanced arrhythmogenicity in the cardiorenal syndrome is unclear. The aim of this study was to characterize electrophysiological and tissue alterations correlated with enhanced arrhythmogenicity in two distinct mouse models of renal failure. METHODS Thirty-week-old 129Sv mice received a high-salt diet and deoxycorticosterone acetate (DOCA) for 8 weeks, followed by an additional period of high-salt diet for 27 weeks (DOCA-salt aged model). Adult CD-1 mice were submitted to 5/6-subtotal nephrectomy (SNx) and treated for 11 weeks with a high-salt diet (SNx-salt adult model). Vulnerability to arrhythmia as well as conduction velocities (CVs) of the hearts were determined ex vivo with epicardial mapping. Subsequently, the hearts were characterized for connexin 43 (Cx43) and fibrosis. RESULTS DOCA-salt and SNx-salt mice developed renal dysfunction characterized by albuminuria. Heart, lung and kidney weights were increased in DOCA-salt mice. Both DOCA-salt and SNx-salt mice were highly susceptible to ventricular arrhythmias. DOCA-salt mice had a significant decrease in both longitudinal and transversal CV in the left ventricle. Histological analysis revealed a significant reduction in Cx43 expression as well as an increase in interstitial fibrosis in both DOCA-salt and SNx-salt mice. CONCLUSION DOCA-salt and SNx-salt treatment induced renal dysfunction, which resulted in structural and electrical cardiac remodeling and enhanced arrhythmogenicity. The reduced Cx43 expression and increased fibrosis levels in these hearts are likely candidates for the formation of the arrhythmogenic substrate.
Collapse
Affiliation(s)
- Magda S C Fontes
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Diana A Papazova
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arianne van Koppen
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sanne de Jong
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sanne M Korte
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lennart G Bongartz
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands ; Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tri Q Nguyen
- Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marti F A Bierhuizen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Harold V M van Rijen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
35
|
Sudden cardiac death in end stage renal disease: unlocking the mystery. J Nephrol 2014; 28:133-41. [PMID: 25391630 DOI: 10.1007/s40620-014-0151-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/09/2014] [Indexed: 01/05/2023]
Abstract
Sudden cardiac death (SCD) is a major cause of concern in end stage renal disease (ESRD), contributing to 70% of cardiovascular mortality and 27% of all-cause mortality in dialysis patients. Yet its mechanisms and pathogenesis remain largely obscure. This review discusses the potential reasons for an exaggerated risk of SCD in ESRD populations taking into account recent studies and registry data and additionally explores the reasons for the reported recent decline in SCD. The types of arrhythmias typical of the hemodialysis population are yet to be fully characterised and in this paper, we introduce an ongoing implantable loop recorder (ILR) based study in hemodialysis patients--CRASH ILR (Cardio Renal Arrhythmia Study in Haemodialysis patients using Implantable Loop Recorders). The findings of this study will hopefully guide the design and implementation of larger ILR based studies before undertaking larger scale interventional therapeutic trials in this high risk population.
Collapse
|