451
|
Anderson AH, Xie D, Wang X, Baudier RL, Orlandi P, Appel LJ, Dember LM, He J, Kusek JW, Lash JP, Navaneethan SD, Ojo A, Rahman M, Roy J, Scialla JJ, Sondheimer JH, Steigerwalt SP, Wilson FP, Wolf M, Feldman HI. Novel Risk Factors for Progression of Diabetic and Nondiabetic CKD: Findings From the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis 2021; 77:56-73.e1. [PMID: 32866540 PMCID: PMC7752839 DOI: 10.1053/j.ajkd.2020.07.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/06/2020] [Indexed: 12/31/2022]
Abstract
RATIONALE & OBJECTIVE Identification of novel risk factors for chronic kidney disease (CKD) progression may inform mechanistic investigations and improve identification of high-risk subgroups. The current study aimed to characterize CKD progression across levels of numerous risk factors and identify independent risk factors for CKD progression among those with and without diabetes. STUDY DESIGN The Chronic Renal Insufficiency Cohort (CRIC) Study is a prospective cohort study of adults with CKD conducted at 7 US clinical centers. SETTING & PARTICIPANTS Participants (N=3,379) had up to 12.3 years of follow-up; 47% had diabetes. PREDICTORS 30 risk factors for CKD progression across sociodemographic, behavioral, clinical, and biochemical domains at baseline. OUTCOMES Study outcomes were estimated glomerular filtration rate (eGFR) slope and the composite of halving of eGFR or initiation of kidney replacement therapy. ANALYTICAL APPROACH Stepwise selection of independent risk factors was performed stratified by diabetes status using linear mixed-effects and Cox proportional hazards models. RESULTS Among those without and with diabetes, respectively, mean eGFR slope was-1.4±3.3 and-2.7±4.7mL/min/1.73m2 per year. Among participants with diabetes, multivariable-adjusted hazard of the composite outcome was approximately 2-fold or greater with higher levels of the inflammatory chemokine CXCL12, the cardiac marker N-terminal pro-B-type natriuretic peptide (NT-proBNP), and the kidney injury marker urinary neutrophil gelatinase-associated lipocalin (NGAL). Among those without diabetes, low serum bicarbonate and higher high-sensitivity troponin T, NT-proBNP, and urinary NGAL levels were all significantly associated with a 1.5-fold or greater rate of the composite outcome. LIMITATIONS The observational study design precludes causal inference. CONCLUSIONS Strong associations for cardiac markers, plasma CXCL12, and urinary NGAL are comparable to that of systolic blood pressure≥140mm Hg, a well-established risk factor for CKD progression. This warrants further investigation into the potential mechanisms that these markers indicate and opportunities to use them to improve risk stratification.
Collapse
Affiliation(s)
- Amanda H Anderson
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| | - Dawei Xie
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Xue Wang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Robin L Baudier
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Paula Orlandi
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Lawrence J Appel
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
| | - Laura M Dember
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - John W Kusek
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - James P Lash
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Sankar D Navaneethan
- Section of Nephrology, Department of Medicine, Baylor University College of Medicine, Houston, TX
| | - Akinlolu Ojo
- University of Kansas School of Medicine, Kansas City, KS
| | - Mahboob Rahman
- Division of Nephrology and Hypertension, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Jason Roy
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ
| | - Julia J Scialla
- Division of Nephrology, Department of Medicine, and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC; Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA; Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA
| | - James H Sondheimer
- Division of Nephrology and Hypertension, Wayne State University School of Medicine, Detroit
| | - Susan P Steigerwalt
- Division of Nephrology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - F Perry Wilson
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Harold I Feldman
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
452
|
El-Achkar TM, Eadon MT, Menon R, Lake BB, Sigdel TK, Alexandrov T, Parikh S, Zhang G, Dobi D, Dunn KW, Otto EA, Anderton CR, Carson JM, Luo J, Park C, Hamidi H, Zhou J, Hoover P, Schroeder A, Joanes M, Azeloglu EU, Sealfon R, Winfree S, Steck B, He Y, D’Agati V, Iyengar R, Troyanskaya OG, Barisoni L, Gaut J, Zhang K, Laszik Z, Rovin BH, Dagher PC, Sharma K, Sarwal MM, Hodgin JB, Alpers CE, Kretzler M, Jain S. A multimodal and integrated approach to interrogate human kidney biopsies with rigor and reproducibility: guidelines from the Kidney Precision Medicine Project. Physiol Genomics 2021; 53:1-11. [PMID: 33197228 PMCID: PMC7847045 DOI: 10.1152/physiolgenomics.00104.2020] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Comprehensive and spatially mapped molecular atlases of organs at a cellular level are a critical resource to gain insights into pathogenic mechanisms and personalized therapies for diseases. The Kidney Precision Medicine Project (KPMP) is an endeavor to generate three-dimensional (3-D) molecular atlases of healthy and diseased kidney biopsies by using multiple state-of-the-art omics and imaging technologies across several institutions. Obtaining rigorous and reproducible results from disparate methods and at different sites to interrogate biomolecules at a single-cell level or in 3-D space is a significant challenge that can be a futile exercise if not well controlled. We describe a "follow the tissue" pipeline for generating a reliable and authentic single-cell/region 3-D molecular atlas of human adult kidney. Our approach emphasizes quality assurance, quality control, validation, and harmonization across different omics and imaging technologies from sample procurement, processing, storage, shipping to data generation, analysis, and sharing. We established benchmarks for quality control, rigor, reproducibility, and feasibility across multiple technologies through a pilot experiment using common source tissue that was processed and analyzed at different institutions and different technologies. A peer review system was established to critically review quality control measures and the reproducibility of data generated by each technology before their being approved to interrogate clinical biopsy specimens. The process established economizes the use of valuable biopsy tissue for multiomics and imaging analysis with stringent quality control to ensure rigor and reproducibility of results and serves as a model for precision medicine projects across laboratories, institutions and consortia.
Collapse
Affiliation(s)
| | | | - Rajasree Menon
- 2University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Blue B. Lake
- 3Jacobs School of Engineering, University of California, San Diego, California
| | - Tara K. Sigdel
- 4University of California San Francisco School of Medicine, San Francisco, California
| | | | - Samir Parikh
- 6Ohio State University College of Medicine, Columbus, Ohio
| | - Guanshi Zhang
- 7UT-Health San Antonio School of Medicine, San Antonio, Texas
| | - Dejan Dobi
- 4University of California San Francisco School of Medicine, San Francisco, California
| | - Kenneth W. Dunn
- 1Indiana University School of Medicine, Indianapolis, Indiana
| | - Edgar A. Otto
- 2University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Christopher R. Anderton
- 7UT-Health San Antonio School of Medicine, San Antonio, Texas,8Pacific Northwest National Laboratory, Richland, Washington
| | - Jonas M. Carson
- 9Schools of Medicine and Public Health, University of Washington, Seattle, Washington
| | - Jinghui Luo
- 2University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Chris Park
- 9Schools of Medicine and Public Health, University of Washington, Seattle, Washington
| | - Habib Hamidi
- 2University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Jian Zhou
- 12Princeton University, Princeton, New Jersey,14Columbia University School of Medicine, New York City, New York
| | - Paul Hoover
- 10Harvard University School of Medicine, Boston Massachusetts
| | - Andrew Schroeder
- 4University of California San Francisco School of Medicine, San Francisco, California
| | - Marianinha Joanes
- 4University of California San Francisco School of Medicine, San Francisco, California
| | | | - Rachel Sealfon
- 12Princeton University, Princeton, New Jersey,14Columbia University School of Medicine, New York City, New York
| | - Seth Winfree
- 1Indiana University School of Medicine, Indianapolis, Indiana
| | - Becky Steck
- 2University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Yongqun He
- 2University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Vivette D’Agati
- 14Columbia University School of Medicine, New York City, New York
| | - Ravi Iyengar
- 11Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Olga G. Troyanskaya
- 12Princeton University, Princeton, New Jersey,14Columbia University School of Medicine, New York City, New York
| | - Laura Barisoni
- 15Duke University School of Medicine, Durham, North Carolina
| | - Joseph Gaut
- 16Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Kun Zhang
- 3Jacobs School of Engineering, University of California, San Diego, California
| | - Zoltan Laszik
- 4University of California San Francisco School of Medicine, San Francisco, California
| | - Brad H. Rovin
- 6Ohio State University College of Medicine, Columbus, Ohio
| | | | - Kumar Sharma
- 7UT-Health San Antonio School of Medicine, San Antonio, Texas
| | - Minnie M. Sarwal
- 4University of California San Francisco School of Medicine, San Francisco, California
| | | | - Charles E. Alpers
- 9Schools of Medicine and Public Health, University of Washington, Seattle, Washington
| | | | - Sanjay Jain
- 16Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
453
|
Hsu PY, Wei YJ, Lee JJ, Niu SW, Huang JC, Hsu CT, Jang TY, Yeh ML, Huang CI, Liang PC, Lin YH, Hsieh MY, Hsieh MH, Chen SC, Dai CY, Lin ZY, Chen SC, Huang JF, Chang JM, Hwang SJ, Chuang WL, Huang CF, Chiu YW, Yu ML. Comedications and potential drug-drug interactions with direct-acting antivirals in hepatitis C patients on hemodialysis. Clin Mol Hepatol 2021; 27:186-196. [PMID: 33317251 PMCID: PMC7820195 DOI: 10.3350/cmh.2020.0180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/20/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/AIMS Direct-acting antivirals (DAAs) have been approved for hepatitis C virus (HCV) treatment in patients with end-stage renal disease (ESRD) on hemodialysis. Nevertheless, the complicated comedications and their potential drug-drug interactions (DDIs) with DAAs might limit clinical practice in this special population. METHODS The number, class, and characteristics of comedications and their potential DDIs with five DAA regimens were analyzed among HCV-viremic patients from 23 hemodialysis centers in Taiwan. RESULTS Of 2,015 hemodialysis patients screened in 2019, 169 patients seropositive for HCV RNA were enrolled (mean age, 65.6 years; median duration of hemodialysis, 5.8 years). All patients received at least one comedication (median number, 6; mean class number, 3.4). The most common comedication classes were ESRD-associated medications (94.1%), cardiovascular drugs (69.8%) and antidiabetic drugs (43.2%). ESRD-associated medications were excluded from DDI analysis. Sofosbuvir/velpatasvir/voxilaprevir had the highest frequency of potential contraindicated DDIs (red, 5.6%), followed by glecaprevir/pibrentasvir (4.0%), sofosbuvir/ledipasvir (1.3%), sofosbuvir/velpatasvir (1.3%), and elbasvir/grazoprevir (0.3%). For potentially significant DDIs (orange, requiring close monitoring or dose adjustments), sofosbuvir/velpatasvir/voxilaprevir had the highest frequency (19.9%), followed by sofosbuvir/ledipasvir (18.2%), glecaprevir/pibrentasvir (12.6%), sofosbuvir/velpatasvir (12.6%), and elbasvir/grazoprevir (7.3%). Overall, lipid-lowering agents were the most common comedication class with red-category DDIs to all DAA regimens (n=62), followed by cardiovascular agents (n=15), and central nervous system agents (n=10). CONCLUSION HCV-viremic patients on hemodialysis had a very high prevalence of comedications with a broad spectrum, which had varied DDIs with currently available DAA regimens. Elbasvir/grazoprevir had the fewest potential DDIs, and sofosbuvir/velpatasvir/voxilaprevir had the most potential DDIs.
Collapse
Affiliation(s)
- Po-Yao Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Ju Wei
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Jung Lee
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheng-Wen Niu
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jiun-Chi Huang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Ting Hsu
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tyng-Yuan Jang
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lun Yeh
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-I Huang
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Cheng Liang
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hung Lin
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Yen Hsieh
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Meng-Hsuan Hsieh
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Preventive Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Szu-Chia Chen
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Preventive Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zu-Yau Lin
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shinn-Cherng Chen
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jer-Ming Chang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shang-Jyh Hwang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Feng Huang
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Division of Hepatobiliary, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine and Hepatitis Research Center, College of Medicine and Center for Cohort Study, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
454
|
Daneshamouz S, Eduok U, Abdelrasoul A, Shoker A. Protein-bound uremic toxins (PBUTs) in chronic kidney disease (CKD) patients: Production pathway, challenges and recent advances in renal PBUTs clearance. NANOIMPACT 2021; 21:100299. [PMID: 35559786 DOI: 10.1016/j.impact.2021.100299] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 06/15/2023]
Abstract
Uremic toxins, a group of uremic retention solutes with high concentration which their accumulation on the body makes several biological problems, have recently gained a large interest. The importance of this issue more targets patients with compromised kidney function since the presence of these toxins in their bodies contributes to serious illness and death. It is reported that around 14% of people are subjected of CKD's problems. Among different classifications of uremic toxins, protein bound uremic toxins are poorly removed from the body as they tightly bind to proteins like serum albumin. A deeper and closer understanding of methods for removing protein bound uremic toxins and their efficiency is of paramount importance. This article discussed the most critical protein bound uremic toxins from different points of view including their chemistry, binding sites, interactions, and their biological impacts. Concerning the toxicity and high concentration, p-cresyl sulfate (PCS), Indoxyl sulfate (IS), 3-Carboxy-4-methyl-5-propyl-2-furanpropionic acid (CMPF), and Indole- 3-acetic acid (IAA) was chosen to study in this article. Results offered that the functional groups of mentioned PBUTs and the way that they interact with the adsorbent play an important role in finding substances for removal of them. Furthermore, the development of nanoparticle (NPs) for promising biomedical purposes has been explored. However, there is still a need for further investigation to find biocompatible substances focusing on the removal of PBUTs. PBUTs are a unique class of uremic toxins whose renal clearance mechanisms and role in uremic pathophysiology are still unclear. This review outlines the biochemical aspects of PBUT/protein binding in a view to explaining their renal formation to elimination mechanisms; some examples are drawn from routes involving albumin-binding with indoxyl sulphate, p-cresyl sulfate, p-cresyl glucuronide and hippuric acid. We have also highlighted the kinetic behaviors during dialytic removal of PBUTs to address future concerns regarding dialytic therapy.
Collapse
Affiliation(s)
- Sana Daneshamouz
- Department of Chemical and Biological Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada
| | - Ubong Eduok
- Department of Chemical and Biological Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada
| | - Amira Abdelrasoul
- Department of Chemical and Biological Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada; Department of Biomedical Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, Saskatchewan S7N 5A9, Canada.
| | - Ahmed Shoker
- Nephrology Division, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK S7N 5E5, Canada; Saskatchewan Transplant Program, St. Paul's Hospital, 1702 20th Street West Saskatoon Saskatchewan S7M 0Z9, Canada
| |
Collapse
|
455
|
Ramick MG, Kirkman DL, Stock JM, Muth BJ, Farquhar WB, Chirinos JA, Doulias PT, Ischiropoulos H, Edwards DG. The effect of dietary nitrate on exercise capacity in chronic kidney disease: a randomized controlled pilot study. Nitric Oxide 2021; 106:17-23. [PMID: 33080411 PMCID: PMC10026360 DOI: 10.1016/j.niox.2020.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/12/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chronic Kidney Disease (CKD) patients exhibit a reduced exercise capacity that impacts quality of life. Dietary nitrate supplementation has been shown to have favorable effects on exercise capacity in disease populations by reducing the oxygen cost of exercise. This study investigated whether dietary nitrates would acutely improve exercise capacity in CKD patients. METHODS AND RESULTS In this randomized, double-blinded crossover study, 12 Stage 3-4 CKD patients (Mean ± SEM: Age, 60 ± 5yrs; eGFR, 50.3 ± 4.6 ml/min/1.73 m2) received an acute dose of 12.6 mmol of dietary nitrate in the form of concentrated beetroot juice (BRJ) and a nitrate depleted placebo (PLA). Skeletal muscle mitochondrial oxidative function was assessed using near-infrared spectroscopy. Cardiopulmonary exercise testing was performed on a cycle ergometer, with intensity increased by 25 W every 3 min until volitional fatigue. Plasma nitric oxide (NO) metabolites (NOm; nitrate, nitrite, low molecular weight S-nitrosothiols, and metal bound NO) were determined by gas-phase chemiluminescence. Plasma NOm values were significantly increased following BRJ (BRJ vs. PLA: 1074.4 ± 120.4 μM vs. 28.4 ± 6.6 μM, p < 0.001). Total work performed (44.4 ± 10.6 vs 39.6 ± 9.9 kJ, p = 0.03) and total exercise time (674 ± 85 vs 627 ± 86s, p = 0.04) were significantly greater following BRJ. Oxygen consumption at the ventilatory threshold was also improved by BRJ (0.90 ± 0.08 vs. 0.74 ± 0.06 L/min, p = 0.04). These changes occurred in the absence of improved skeletal muscle mitochondrial oxidative capacity (p = 0.52) and VO2peak (p = 0.35). CONCLUSIONS Our findings demonstrate that inorganic nitrate can acutely improve exercise capacity in CKD patients. The effects of chronic nitrate supplementation on CKD related exercise intolerance should be investigated in future studies.
Collapse
Affiliation(s)
- Meghan G Ramick
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA; Department of Kinesiology, West Chester University, West Chester, PA, USA
| | - Danielle L Kirkman
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA; Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Joseph M Stock
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA
| | - Bryce J Muth
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA; School of Health Sciences, Stockton University, Stockton, NJ, USA
| | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA
| | - Julio A Chirinos
- Division of Cardiovascular Medicine. Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paschalis-Thomas Doulias
- Laboratory of Biochemistry, Department of Chemistry, School of Sciences, University of Ioannina, Ioannina, 45110, Greece; Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Harry Ischiropoulos
- Laboratory of Biochemistry, Department of Chemistry, School of Sciences, University of Ioannina, Ioannina, 45110, Greece; Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, USA.
| |
Collapse
|
456
|
Thiem U, Buxhofer‐Ausch V, Kranewitter W, Webersinke G, Enkner W, Cejka D. Successful kidney transplantation in a patient with pre-existing chronic myeloid leukemia treated with imatinib. Am J Transplant 2021; 21:405-409. [PMID: 32654389 PMCID: PMC7818412 DOI: 10.1111/ajt.16194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/29/2020] [Accepted: 06/28/2020] [Indexed: 01/25/2023]
Abstract
Active malignancy is an absolute contraindication to kidney transplantation. As for chronic myeloid leukemia (CML), a Philadelphia chromosome-positive myeloproliferative neoplasm, the introduction of tyrosine kinase inhibitors has transformed CML from a lethal into a manageable chronic disease with a close-to-normal life expectancy. To date it is unknown whether kidney transplantation can be safely performed in patients with pre-existing CML. We describe the clinical course of a 57-year-old male patient with chronic kidney disease caused by reflux nephropathy. This patient had undergone first kidney transplantation 20 years earlier and had again been on chronic hemodialysis for 6 years when CML was diagnosed. First-line therapy with 400 mg imatinib daily was well tolerated and induced an optimal cytogenetic and molecular response 3 months after initiation. One and a half years after CML diagnosis, a second kidney transplantation from a deceased donor was performed. Immunosuppression included basiliximab, tacrolimus, mycophenolate mofetil, and corticosteroids. Currently, 2 years posttransplant, renal allograft function is stable (serum creatinine 1.09 mg/dL, estimated glomerular filtration rate 75 mL/min per 1.73 m2 ), and CML remains in deep molecular remission with imatinib. Imatinib-treated CML in deep molecular remission could be regarded as inactive malignancy and may therefore not be viewed as an absolute contraindication to kidney transplantation.
Collapse
Affiliation(s)
- Ursula Thiem
- Department of Nephrology and Hypertension DiseasesTransplantation Medicine and RheumatologyOrdensklinikum LinzKrankenhaus ElisabethinenLinzAustria,Faculty of MedicineJohannes Kepler University LinzLinzAustria
| | - Veronika Buxhofer‐Ausch
- Department of Internal Medicine I: Hematology with Stem Cell Transplantation, Hemostaseology and Medical OncologyOrdensklinikum LinzKrankenhaus ElisabethinenLinzAustria
| | - Wolfgang Kranewitter
- Laboratory for Molecular Biology and Tumor CytogeneticsDepartment of Internal Medicine IOrdensklinikum LinzBarmherzige SchwesternLinzAustria
| | - Gerald Webersinke
- Laboratory for Molecular Biology and Tumor CytogeneticsDepartment of Internal Medicine IOrdensklinikum LinzBarmherzige SchwesternLinzAustria
| | - Wolfgang Enkner
- Department of Nephrology and Hypertension DiseasesTransplantation Medicine and RheumatologyOrdensklinikum LinzKrankenhaus ElisabethinenLinzAustria
| | - Daniel Cejka
- Department of Nephrology and Hypertension DiseasesTransplantation Medicine and RheumatologyOrdensklinikum LinzKrankenhaus ElisabethinenLinzAustria
| |
Collapse
|
457
|
Portales-Castillo I, Yee J, Tanaka H, Fenves AZ. Beta-2 Microglobulin Amyloidosis: Past, Present, and Future. KIDNEY360 2020; 1:1447-1455. [PMID: 35372889 DOI: 10.34067/kid.0004922020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/21/2020] [Indexed: 11/27/2022]
Abstract
Almost half a century has elapsed since the first description of dialysis-related amyloidosis (DRA), a disorder caused by excessive accumulation of β-2 microglobulin (B2M). Within that period, substantial advances in RRT occurred. These improvements have led to a decrease in the incidence of DRA. In many countries, DRA is considered a "disappearing act" or complication. Although the prevalence of patients living with RRT increases, not all will have access to kidney transplantation. Consequently, the number of patients requiring interventions for treatment of DRA is postulated to increase. This postulate has been borne out in Japan, where the number of patients with ESKD requiring surgery for carpal tunnel continues to increase. Clinicians treating patients with ESKD have treatment options to improve B2M clearance; however, there is a need to identify ways to translate improved B2M clearance into improved quality of life for patients undergoing long-term dialysis.
Collapse
Affiliation(s)
- Ignacio Portales-Castillo
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jerry Yee
- Division of Nephrology and Hypertension, Henry Ford Hospital, Detroit, Michigan
| | - Hiroshi Tanaka
- Division of Nephrology, Department of Medicine, Mihara Red Cross Hospital, Mihara, Japan
| | - Andrew Z Fenves
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
458
|
Schunk SJ, Speer T, Fliser D. Heart and kidney disease: a cardiovascular high-risk constellation. Herz 2020; 46:206-211. [PMID: 33377985 DOI: 10.1007/s00059-020-05012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 11/30/2022]
Abstract
Chronic kidney disease (CKD) is associated with substantial cardiovascular morbidity and mortality. This is mediated by highly prevalent traditional cardiovascular risk factors such as arterial hypertension and diabetes mellitus in patients with CKD, but also by the presence of CKD-specific so-called nontraditional cardiovascular risk factors such as vascular calcification, uremic toxins, uremic dyslipidemia, inflammation, and oxidative stress. Therefore, the primary and secondary prevention of cardiovascular disease represents an important part of the care of patients with CKD. This entails optimal control of blood pressure and diabetes, treatment of the uremic dyslipidemia, as well as life-style modifying factors such as weight reduction and smoking cessation.
Collapse
Affiliation(s)
- Stefan J Schunk
- Klinik für Innere Medizin IV, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany. .,Klinik für Innere Medizin IV, Nieren- und Hochdruckkrankheiten, Universitätsklinikum des Saarlandes, Kirrberger Straße, Gebäude 41, 66421, Homburg/Saar, Germany.
| | - Thimoteus Speer
- Klinik für Innere Medizin IV, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany.,Translationale Kardio-Renale Medizin, Universität des Saarlandes, Homburg/Saar, Germany
| | - Danilo Fliser
- Klinik für Innere Medizin IV, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| |
Collapse
|
459
|
Murakami N, Mulvaney P, Danesh M, Abudayyeh A, Diab A, Abdel-Wahab N, Abdelrahim M, Khairallah P, Shirazian S, Kukla A, Owoyemi IO, Alhamad T, Husami S, Menon M, Santeusanio A, Blosser CD, Zuniga SC, Soler MJ, Moreso F, Mithani Z, Ortiz-Melo D, Jaimes EA, Gutgarts V, Lum E, Danovitch GM, Cardarelli F, Drews RE, Bassil C, Swank JL, Westphal S, Mannon RB, Shirai K, Kitchlu A, Ong S, Machado SM, Mothi SS, Ott PA, Rahma O, Hodi FS, Sise ME, Gupta S, Leaf DE, Devoe CE, Wanchoo R, Nair VV, Schmults CD, Hanna GJ, Sprangers B, Riella LV, Jhaveri KD. A multi-center study on safety and efficacy of immune checkpoint inhibitors in cancer patients with kidney transplant. Kidney Int 2020; 100:196-205. [PMID: 33359528 DOI: 10.1016/j.kint.2020.12.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are widely used for various malignancies. However, their safety and efficacy in patients with a kidney transplant have not been defined. To delineate this, we conducted a multicenter retrospective study of 69 patients with a kidney transplant receiving ICIs between January 2010 and May 2020. For safety, we assessed the incidence, timing, and risk factors of acute graft rejection. For efficacy, objective response rate and overall survival were assessed in cutaneous squamous cell carcinoma and melanoma, the most common cancers in our cohort, and compared with stage-matched 23 patients with squamous cell carcinoma and 14 with melanoma with a kidney transplant not receiving ICIs. Following ICI treatment, 29 out of 69 (42%) patients developed acute rejection, 19 of whom lost their allograft, compared with an acute rejection rate of 5.4% in the non-ICI cohort. Median time from ICI initiation to rejection was 24 days. Factors associated with a lower risk of rejection were mTOR inhibitor use (odds ratio 0.26; 95% confidence interval, 0.09-0.72) and triple-agent immunosuppression (0.67, 0.48-0.92). The objective response ratio was 36.4% and 40% in the squamous cell carcinoma and melanoma subgroups, respectively. In the squamous cell carcinoma subgroup, overall survival was significantly longer in patients treated with ICIs (median overall survival 19.8 months vs. 10.6 months), whereas in the melanoma subgroup, overall survival did not differ between groups. Thus, ICIs were associated with a high risk of rejection in patients with kidney transplants but may lead to improved cancer outcomes. Prospective studies are needed to determine optimal immunosuppression strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Naoka Murakami
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - Patrick Mulvaney
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Melissa Danesh
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ala Abudayyeh
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Adi Diab
- Department of Melanoma Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Noha Abdel-Wahab
- Department of Melanoma Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of General Internal Medicine, Section of Rheumatology and Clinical Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Maen Abdelrahim
- Institute for Academic Medicine and Weill Cornell Medical College, Houston Methodist Cancer Center, Houston, Texas, USA
| | - Pascale Khairallah
- Division of Nephrology, Columbia University Medical Center, New York, New York, USA
| | - Shayan Shirazian
- Division of Nephrology, Columbia University Medical Center, New York, New York, USA
| | - Aleksandra Kukla
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Itunu O Owoyemi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tarek Alhamad
- Division of Nephrology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Samir Husami
- Division of Nephrology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Madhav Menon
- Division of Nephrology, Recanati Millar Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew Santeusanio
- Division of Nephrology, Recanati Millar Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Maria Jose Soler
- Nephrology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Francesc Moreso
- Nephrology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Zain Mithani
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David Ortiz-Melo
- Division of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Edgar A Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center and Weil Cornell Medical College, New York, New York, USA
| | - Victoria Gutgarts
- Renal Service, Memorial Sloan Kettering Cancer Center and Weil Cornell Medical College, New York, New York, USA
| | - Erik Lum
- Division of Nephrology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Gabriel M Danovitch
- Division of Nephrology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Francesca Cardarelli
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Reed E Drews
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Claude Bassil
- Division of Nephrology and Hypertension, University of South Florida, Tampa, Florida; Renal Service, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Jennifer L Swank
- Division of Nephrology and Hypertension, University of South Florida, Tampa, Florida; Renal Service, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Scott Westphal
- Division of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Roslyn B Mannon
- Division of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Keisuke Shirai
- Department of Hematology-Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Abhijat Kitchlu
- Division of Nephrology, University Health Network, University of Toronto, Ontario, Canada
| | - Song Ong
- Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shana M Machado
- Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suraj S Mothi
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Osama Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Meghan E Sise
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Craig E Devoe
- Division of Medical Oncology and Hematology, Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York, USA
| | - Rimda Wanchoo
- Division of Kidney Disease and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Vinay V Nair
- Division of Kidney Disease and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | | | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ben Sprangers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Belgium; Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Leonardo V Riella
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kenar D Jhaveri
- Division of Kidney Disease and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.
| |
Collapse
|
460
|
Alphonse S, Polichnowski AJ, Griffin KA, Bidani AK, Williamson GA. Autoregulatory Efficiency Assessment in Kidneys Using Deep Learning. PROCEEDINGS OF THE ... EUROPEAN SIGNAL PROCESSING CONFERENCE (EUSIPCO). EUSIPCO (CONFERENCE) 2020; 2020:1165-1169. [PMID: 38288370 PMCID: PMC10824283 DOI: 10.23919/eusipco47968.2020.9287447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
A convolutional deep neural network is employed to assess renal autoregulation using time series of arterial blood pressure and blood flow rate measurements in conscious rats. The network is trained using representative data samples from rats with intact autoregulation and rats whose autoregulation is impaired by the calcium channel blocker amlodipine. Network performance is evaluated using test data of the types used for training, but also with data from other models for autoregulatory impairment, including different calcium channel blockers and also renal mass reduction. The network is shown to provide effective classification for impairments from calcium channel blockers. However, the assessment of autoregulation when impaired by renal mass reduction was not as clear, evidencing a different signature in the hemodynamic data for that impairment model. When calcium channel blockers were given to those animals, however, the classification again was effective.
Collapse
Affiliation(s)
- Sebastian Alphonse
- Dept. of Elec. and Comp. Engr., Illinois Institute of Technology Chicago, IL, U.S.A
| | - Aaron J Polichnowski
- Department of Biomedical Sciences East Tennessee State University, Johnson City, TN, U.S.A
| | - Karen A Griffin
- Departments of Medicine Loyola Univ. Med. Ctr. and Edward Hines, Jr. VA Hosp. Maywood, IL, U.S.A
| | - Anil K Bidani
- Departments of Medicine Loyola Univ. Med. Ctr. and Edward Hines, Jr. VA Hosp. Maywood, IL, U.S.A
| | | |
Collapse
|
461
|
Cansby E, Caputo M, Gao L, Kulkarni NM, Nerstedt A, Ståhlman M, Borén J, Porosk R, Soomets U, Pedrelli M, Parini P, Marschall HU, Nyström J, Howell BW, Mahlapuu M. Depletion of protein kinase STK25 ameliorates renal lipotoxicity and protects against diabetic kidney disease. JCI Insight 2020; 5:140483. [PMID: 33170807 PMCID: PMC7819747 DOI: 10.1172/jci.insight.140483] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetic kidney disease (DKD) is the most common cause of severe renal disease worldwide and the single strongest predictor of mortality in diabetes patients. Kidney steatosis has emerged as a critical trigger in the pathogenesis of DKD; however, the molecular mechanism of renal lipotoxicity remains largely unknown. Our recent studies in genetic mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine protein kinase 25 (STK25) as a critical regulator of ectopic lipid storage in several metabolic organs prone to diabetic damage. Here, we demonstrate that overexpression of STK25 aggravates renal lipid accumulation and exacerbates structural and functional kidney injury in a mouse model of DKD. Reciprocally, inhibiting STK25 signaling in mice ameliorates diet-induced renal steatosis and alleviates the development of DKD-associated pathologies. Furthermore, we find that STK25 silencing in human kidney cells protects against lipid deposition, as well as oxidative and endoplasmic reticulum stress. Together, our results suggest that STK25 regulates a critical node governing susceptibility to renal lipotoxicity and that STK25 antagonism could mitigate DKD progression.
Collapse
Affiliation(s)
| | - Mara Caputo
- Department of Chemistry and Molecular Biology and
| | - Lei Gao
- Department of Chemistry and Molecular Biology and
| | | | | | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rando Porosk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Ursel Soomets
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | | | - Paolo Parini
- Department of Laboratory Medicine and.,Metabolism Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Brian W Howell
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | | |
Collapse
|
462
|
Li WY, Chiu FC, Zeng JK, Li YW, Huang SH, Yeh HC, Cheng BW, Yang FJ. Mobile Health App With Social Media to Support Self-Management for Patients With Chronic Kidney Disease: Prospective Randomized Controlled Study. J Med Internet Res 2020; 22:e19452. [PMID: 33320101 PMCID: PMC7772070 DOI: 10.2196/19452] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/12/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a global health burden. Self-management plays a key role in improving modifiable risk factors. OBJECTIVE The aim of this study was to evaluate the effectiveness of wearable devices, a health management platform, and social media at improving the self-management of CKD, with the goal of establishing a new self-management intervention model. METHODS In a 90-day prospective experimental study, a total of 60 people with CKD at stages 1-4 were enrolled in the intervention group (n=30) and control group (n=30). All participants were provided with wearable devices that collected exercise-related data. All participants maintained dietary diaries using a smartphone app. All dietary and exercise information was then uploaded to a health management platform. Suggestions about diet and exercise were provided to the intervention group only, and a social media group was created to inspire the participants in the intervention group. Participants' self-efficacy and self-management questionnaire scores, Kidney Disease Quality of Life scores, body composition, and laboratory examinations before and after the intervention were compared between the intervention and control groups. RESULTS A total of 49 participants completed the study (25 in the intervention group and 24 in the control group); 74% of the participants were men and the mean age was 51.22 years. There were no differences in measured baseline characteristics between the groups except for educational background. After the intervention, the intervention group showed significantly higher scores for self-efficacy (mean 171.28, SD 22.92 vs mean 142.21, SD 26.36; P<.001) and self-management (mean 54.16, SD 6.71 vs mean 47.58, SD 6.42; P=.001). Kidney Disease Quality of Life scores were also higher in the intervention group (mean 293.16, SD 34.21 vs mean 276.37, SD 32.21; P=.02). The number of steps per day increased in the intervention group (9768.56 in week 1 and 11,389.12 in week 12). The estimated glomerular filtration rate (eGFR) of the intervention group was higher than that of the control group (mean 72.47, SD 24.28 vs mean 59.69, SD 22.25 mL/min/1.73m2; P=.03) and the decline in eGFR was significantly slower in the intervention group (-0.56 vs -4.58 mL/min/1.73m2). There were no differences in body composition between groups postintervention. CONCLUSIONS The use of wearable devices, a health management platform, and social media support not only strengthened self-efficacy and self-management but also improved quality of life and a slower eGFR decline in people with CKD at stages 1-4. These results outline a new self-management model to promote healthy lifestyle behaviors for patients with CKD. TRIAL REGISTRATION ClinicalTrials.gov NCT04617431; https://www.clinicaltrials.gov/ct2/show/NCT04617431.
Collapse
Affiliation(s)
- Wen-Yi Li
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Yun Lin Branch, Douliu, Taiwan
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fu-Chun Chiu
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Cardiovascular Division, Department of Internal Medicine, National Taiwan University Hospital Yun Lin Branch, Douliu, Taiwan
| | - Jyun-Kai Zeng
- Department of Industrial Engineering and Management, National Yunlin University of Science and Technology, Douliu, Taiwan
| | - Yao-Wei Li
- Department of Industrial Engineering and Management, National Yunlin University of Science and Technology, Douliu, Taiwan
| | - Su-Hua Huang
- Department of Dietetics, National Taiwan University Hospital Yun Lin Branch, Douliu, Taiwan
| | - Hui-Chin Yeh
- Department of Industrial Engineering and Management, National Yunlin University of Science and Technology, Douliu, Taiwan
- Department of Applied Foreign Languages, National Yunlin University of Science and Technology, Douliu, Taiwan
| | - Bor-Wen Cheng
- Department of Industrial Engineering and Management, National Yunlin University of Science and Technology, Douliu, Taiwan
| | - Feng-Jung Yang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Yun Lin Branch, Douliu, Taiwan
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
463
|
van Rijn MH, Alencar de Pinho N, Wetzels JF, van den Brand JA, Stengel B. Worldwide Disparity in the Relation Between CKD Prevalence and Kidney Failure Risk. Kidney Int Rep 2020; 5:2284-2291. [PMID: 33305122 PMCID: PMC7710841 DOI: 10.1016/j.ekir.2020.09.040] [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: 03/16/2020] [Revised: 08/21/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction The incidence of kidney replacement therapy (KRT) for kidney failure varies internationally much more than chronic kidney disease (CKD) prevalence. This ecologic study investigated the relation of CKD prevalence to KRT and mortality risks by world region. Methods We used data from Global Burden of Disease and KRT registries worldwide with linear models to estimate the percentages of variance in KRT incidence and all-cause mortality explained by age-adjusted prevalence of CKD stages 3 to 5, overall and by gender, in 61 countries classified in 3 regions: high income (n = 28), Eastern and Central Europe (n = 15), and other (n = 18). Results The incidence of KRT ranged from 89 to 378 per million population in high-income regions, 32 to 222 per million population in Central and Eastern Europe, and 22 to 493 per million population in the other region; age-adjusted CKD prevalence ranged from 5.5% to 10.4%, 7.6% to 13.7%, and 7.4% to 13.1%, respectively. The relation between these indicators was positive in high-income countries, negative in Central and Eastern Europe, and null in the other region. Age-adjusted CKD prevalence explained 40% of the variance in KRT incidence (P < 0.001) in high-income countries. The explained variance of age-adjusted mortality was close to 0 in high-income countries and positive at 19% (P = 0.10) in Central and Eastern Europe and at 11% (P = 0.17) in the other region. Results were consistent by gender. Conclusion This study raises awareness on the significant part of the gaps in KRT incidence across countries not explained by the number of individuals with CKD, even in high-income countries where access to KRT is not limited.
Collapse
Affiliation(s)
- Marieke H.C. van Rijn
- Department of Nephrology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Université Paris-Saclay, Université de Versailles-Saint-Quentin-en-Yvelines, University Paris Sud, INSERM, Clinical Epidemiology Team, Centre for Research in Epidemiology and Population Health, Villejuif, France
| | - Natalia Alencar de Pinho
- Université Paris-Saclay, Université de Versailles-Saint-Quentin-en-Yvelines, University Paris Sud, INSERM, Clinical Epidemiology Team, Centre for Research in Epidemiology and Population Health, Villejuif, France
- Correspondence: Natalia Alencar de Pinho, Clinical Epidemiology Team, Centre for Research in Epidemiology and Population Health, INSERM U1018, 16, avenue Paul Vaillant Couturier, 94807 Villejuif, France.
| | - Jack F. Wetzels
- Department of Nephrology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan A.J.G. van den Brand
- Department of Nephrology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Benedicte Stengel
- Université Paris-Saclay, Université de Versailles-Saint-Quentin-en-Yvelines, University Paris Sud, INSERM, Clinical Epidemiology Team, Centre for Research in Epidemiology and Population Health, Villejuif, France
| |
Collapse
|
464
|
Muscat P, Weinman J, Farrugia E, Camilleri L, Chilcot J. Illness perceptions predict mortality in patients with predialysis chronic kidney disease: a prospective observational study. BMC Nephrol 2020; 21:537. [PMID: 33302894 PMCID: PMC7727218 DOI: 10.1186/s12882-020-02189-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 11/26/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Illness perceptions have been shown to predict a range of psychosocial and clinical outcomes in kidney disease; including quality of life, distress, treatment adherence and even survival in end-stage renal disease patients on dialysis. The aim of this study was to evaluate whether illness perceptions impact mortality in incident predialysis Chronic Kidney Disease (CKD) patients. METHODS Over the study period between September 2015 and June 2019, a total of 200 participants with predialysis CKD were recruited from the Nephrology Outpatient's clinics at Mater Dei Hospital, Malta. The participants were followed up until June 2019, and the mortality information was collected. Cox proportional hazards models were used to examine the association between illness perceptions, and mortality risk, after adjustment for covariates including distress, kidney function, co-morbidity and psychological distress. RESULTS Of the 200 cases available for analysis, there were 43 deaths. The mean survival time was 718.55 days (min. 3 days, max. 1297 days). The cumulative survival 1-year post the assessment of the Revised Illness Perceptions Questionnaire (IPQ-R) was 93%. Stronger identity beliefs (HR = 1.199, 95% CI: 1.060-1.357, p = 0.004), perceptions of a chronic timeline (HR = 1.065, 95% CI: 1.003-1.132, p = 0.041), personal control beliefs (HR = 0.845, 95% CI: 0.748-0.955, p = 0.007) and perceptions of control over the treatment (HR = 0.812, 95% CI: 0.725-0.909, p = 0.000) demonstrated a significant association with mortality after controlling covariates. In a subsequent saturated model, perceived identity, chronic timeline and treatment control perceptions remained significant predictors of mortality, together with serum albumin, comorbidities and urea. CONCLUSIONS CKD patients' perceptions of treatment control, perceptions of a chronic timeline and perceived illness identity predict survival independently of clinical prognostic factors, including kidney function and co-morbidity. Illness perceptions are important and potentially modifiable risk factors in CKD. Further studies are required to test whether the assessment and the implementation of psychological interventions aimed to modify maladaptive illness perceptions influence clinical outcomes in CKD.
Collapse
Affiliation(s)
- Priscilla Muscat
- Renal Unit, Nephrology Department, Mater Dei Hospital, Msida, MSD 2090, Malta.
- Health Psychology Section, Psychology Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5th Floor Bermondsey Wing, Guy's Campus, London Bridge, London, SE1 9RT, UK.
| | - John Weinman
- Institute of Pharmaceutical Science, Pharmaceutical Sciences Clinical Academic Group King's College London, 5th floor, Franklin -Wilkins Building. 150 Stamford Street, London, SE19NH, UK
| | - Emanuel Farrugia
- Renal Unit, Nephrology Department, Mater Dei Hospital, Msida, MSD 2090, Malta
| | - Liberato Camilleri
- Statistics and Operations Research Department University of Malta, Msida, MSD 2080, Malta
| | - Joseph Chilcot
- Health Psychology Section, Psychology Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5th Floor Bermondsey Wing, Guy's Campus, London Bridge, London, SE1 9RT, UK.
| |
Collapse
|
465
|
Kirkman DL, Bohmke N, Carbone S, Garten RS, Rodriguez-Miguelez P, Franco RL, Kidd JM, Abbate A. Exercise intolerance in kidney diseases: physiological contributors and therapeutic strategies. Am J Physiol Renal Physiol 2020; 320:F161-F173. [PMID: 33283641 DOI: 10.1152/ajprenal.00437.2020] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Exertional fatigue, defined as the overwhelming and debilitating sense of sustained exhaustion that impacts the ability to perform activities of daily living, is highly prevalent in chronic kidney disease (CKD) and end-stage renal disease (ESRD). Subjective reports of exertional fatigue are paralleled by objective measurements of exercise intolerance throughout the spectrum of the disease. The prevalence of exercise intolerance is clinically noteworthy, as it leads to increased frailty, worsened quality of life, and an increased risk of mortality. The physiological underpinnings of exercise intolerance are multifaceted and still not fully understood. This review aims to provide a comprehensive outline of the potential physiological contributors, both central and peripheral, to kidney disease-related exercise intolerance and highlight current and prospective interventions to target this symptom. In this review, the CKD-related metabolic derangements, cardiac and pulmonary dysfunction, altered physiological responses to oxygen consumption, vascular derangements, and sarcopenia are discussed in the context of exercise intolerance. Lifestyle interventions to improve exertional fatigue, such as aerobic and resistance exercise training, are discussed, and the lack of dietary interventions to improve exercise tolerance is highlighted. Current and prospective pharmaceutical and nutraceutical strategies to improve exertional fatigue are also broached. An extensive understanding of the pathophysiological mechanisms of exercise intolerance will allow for the development of more targeted therapeutic approached to improve exertional fatigue and health-related quality of life in CKD and ESRD.
Collapse
Affiliation(s)
- Danielle L Kirkman
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Natalie Bohmke
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Salvatore Carbone
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia.,Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Ryan S Garten
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Paula Rodriguez-Miguelez
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Robert L Franco
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Jason M Kidd
- Division of Nephrology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia.,Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
466
|
Diamond LH, Armistead NC, Lupu DE, Moss AH. Recommendations for Public Policy Changes to Improve Supportive Care for Seriously Ill Patients With Kidney Disease. Am J Kidney Dis 2020; 77:529-537. [PMID: 33278476 DOI: 10.1053/j.ajkd.2020.09.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/29/2020] [Indexed: 01/03/2023]
Abstract
National and international nephrology organizations have identified substantial unmet supportive care needs of patients with kidney disease and issued recommendations. In the United States, the most recent comprehensive effort to change kidney care, the Advancing American Kidney Health Initiative, does not explicitly address supportive care needs, although it attempts to implement more patient-centered care. This Perspective from the leaders of the Coalition for Supportive Care of Kidney Patients advocates for urgent policy changes to improve patient-centered care and the quality of life of seriously ill patients with kidney disease. It argues for the provision of supportive care by an interdisciplinary team led by nephrology clinicians to improve shared decision-making, advance care planning, pain and symptom management, the explicit offering of active medical management without dialysis as an option for patients who may not benefit from dialysis, and the removal by the Centers for Medicare & Medicaid Services and all other payors of financial and regulatory disincentives to quality supportive care, including hospice, for patients with or approaching kidney failure. It also emphasizes that all educational and accreditation programs for nephrology clinicians include kidney supportive care and its essential role in the care of patients with kidney disease.
Collapse
Affiliation(s)
- Louis H Diamond
- Center for Aging, Health and Humanities, George Washington University School of Nursing, Washington, DC; Coalition for Supportive Care of Kidney Patients, Washington, DC
| | - Nancy C Armistead
- Center for Aging, Health and Humanities, George Washington University School of Nursing, Washington, DC; Coalition for Supportive Care of Kidney Patients, Washington, DC
| | - Dale E Lupu
- Center for Aging, Health and Humanities, George Washington University School of Nursing, Washington, DC; Coalition for Supportive Care of Kidney Patients, Washington, DC
| | - Alvin H Moss
- Coalition for Supportive Care of Kidney Patients, Washington, DC; Center for Health Ethics and Law, West Virginia University School of Medicine, Morgantown, WV.
| | | |
Collapse
|
467
|
Chia JMX, Goh ZS, Seow PS, Seow TYY, Choo JCJ, Foo MWY, Newman S, Griva K. Psychosocial Factors, Intentions to Pursue Arteriovenous Dialysis Access, and Access Outcomes: A Cohort Study. Am J Kidney Dis 2020; 77:931-940. [PMID: 33279557 DOI: 10.1053/j.ajkd.2020.09.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 09/17/2020] [Indexed: 11/11/2022]
Abstract
RATIONALE & OBJECTIVE Suboptimal dialysis preparation of patients with chronic kidney disease (CKD) is common, but little is known about its relationship to psychosocial factors. This study aimed to assess patients' attitudes about access creation and to identify factors associated with patients' intentions regarding dialysis access creation and outcomes. STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS 190 patients with stage 4/5 CKD not receiving dialysis treated at 2 hospitals in Singapore and 128 of their family members. PREDICTORS Self-reported measures of illness perception, health-related quality of life, and attitudes toward access creation. Sociodemographic and clinical measures were also obtained. OUTCOME Intention to create an arteriovenous fistula (AVF; ie, proceed with access vs wait and see) and time to creation of a functional AVF. ANALYTICAL APPROACH Exploratory factor analysis (EFA) was undertaken to construct internally consistent subscales for a newly developed questionnaire about attitudes toward access creation. Logistic regression and cause-specific hazards models were conducted to identify psychosocial factors associated with patients' access creation intentions and access outcomes, respectively. RESULTS EFA (explained 50.1% variance) revealed 4 domains: access and dialysis concerns, need for dialysis, worry about cost, and value of access. A high risk of intention to delay access creation (51.1%) was found among patients despite early referral and education. Multivariable analysis (R2=0.45) showed that the intention to proceed with access creation was associated with greater perceived value from access (odds ratio, 2.61; 95% CI, 1.46-4.65; P<0.001). LIMITATIONS Limited generalization, as only those already receiving nephrology care were studied. CONCLUSIONS Approximately half of the patients studied planned to delay access creation. The questionnaire developed to evaluate attitudes about access creation may help identify individuals for whom decision-support programs would be useful. These findings highlight the need to understand and address patients' concerns about access creation.
Collapse
Affiliation(s)
- Jace Ming Xuan Chia
- Centre for Population Health Sciences, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Zhong Sheng Goh
- Centre for Population Health Sciences, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Pei Shing Seow
- Department of General Medicine, Khoo Teck Puat Hospital, Singapore
| | | | | | | | - Stanton Newman
- School of Health Sciences, Division of Health Services Research and Management, City University of London, London, United Kingdom
| | - Konstadina Griva
- Centre for Population Health Sciences, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore.
| |
Collapse
|
468
|
Boyle SM, Zhao Y, Chou E, Moore K, Harhay MN. Neighborhood context and kidney disease in Philadelphia. SSM Popul Health 2020; 12:100646. [PMID: 32939392 PMCID: PMC7476869 DOI: 10.1016/j.ssmph.2020.100646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 01/10/2023] Open
Abstract
Neighborhood context might influence the risk of chronic kidney disease (CKD), a condition that impacts approximately 10% of the United States population and is associated with significant morbidity, mortality, and costs. We included a sample of 23,692 individuals in Philadelphia, Pennsylvania, who were seen in a large academic primary care practice between January 1, 2016 and December 31, 2017. We used generalized linear equations to estimate the associations between indicators of neighborhood context (e.g., proximity to healthy foods stores, neighborhood walkability, social capital, crime rate, socioeconomic status) and CKD, adjusted for age, sex, race/ethnicity, and insurance coverage. Among those with CKD, secondary outcomes were poor glycemic control (hemoglobin A1c ≥ 6.5%) and uncontrolled blood pressure (systolic ≥ 140 mm Hg and/or diastolic ≥ 90 mm Hg). The cohort represented residents from 97% of Philadelphia census tracts. CKD prevalence was 10%. When all neighborhood context metrics were considered collectively, only lower neighborhood socioeconomic index (a composite assessment of neighborhood income, educational attainment, and occupation) was associated with a higher risk of CKD (lowest tertile vs. highest tertile: adjusted relative risk [aRR] 1.46 [1.25, 1.69]; mid-tertile vs. highest-tertile: aRR 1.35 [1.25, 1.52]). Among those with CKD, compared to residence in the most walkable neighborhoods (i.e., where most essential resources are accessible by foot), residence in neighborhoods with mid-level WalkScore® (i.e., where only some essential neighborhood resources are accessible by foot) was independently associated with poor glycemic control (aRR 1.20, 95% CI 1.01-1.42). These findings suggest a potential role for measures of neighborhood socioeconomic status in identifying communities that would benefit from screening and treatment for CKD. Studies are also needed to determine mechanisms to explain why residence in neighborhoods not easily navigated by foot or car might hinder glycemic control among people with CKD.
Collapse
Affiliation(s)
- Suzanne M. Boyle
- Department of Medicine, Section of Nephrology, Hypertension and Kidney Transplantation, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Yuzhe Zhao
- Urban Health Collaborative, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Edgar Chou
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Kari Moore
- Urban Health Collaborative, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Meera N. Harhay
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
- Tower Health Transplant Institute, Tower Health System, West Reading, Pennsylvania, USA
| |
Collapse
|
469
|
Hsu CY, Parikh RV, Pravoverov LN, Zheng S, Glidden DV, Tan TC, Go AS. Implication of Trends in Timing of Dialysis Initiation for Incidence of End-stage Kidney Disease. JAMA Intern Med 2020; 180:1647-1654. [PMID: 33044519 PMCID: PMC7551228 DOI: 10.1001/jamainternmed.2020.5009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE In the last 2 decades, there have been notable changes in the level of estimated glomerular filtration rate (eGFR) at which patients initiate long-term dialysis in the US and around the world. How changes over time in the likelihood of dialysis initiation at any given eGFR level in at-risk patients are associated with the population burden of end-stage kidney disease (ESKD) has not been not well defined. OBJECTIVE To examine temporal trends in long-term dialysis initiation by level of eGFR and to quantify how these patterns are associated with the number of patients with ESKD. DESIGN, SETTING, AND PARTICIPANTS Retrospective cohort study analyzing data obtained from a large, integrated health care delivery system in Northern California from 2001 to 2018 in successive 3-year intervals. Included individuals, ranging in number from as few as 983 122 (2001-2003) to as many as 1 844 317 (2016-2018), were adult members with 1 or more outpatient serum creatinine levels determined in the prior year. MAIN OUTCOMES AND MEASURES One-year risk of initiating long-term dialysis stratified by eGFR levels. Multivariable logistic regression was performed to assess temporal trends in each 3-year cohort with adjustment for age, sex, race, and diabetes status. The potential change in dialysis initiation in the final cohort (2016-2018) was estimated using the relative difference between the standardized risks in the initial cohort (2001-2003) and the final cohort. RESULTS In the initial 3-year cohort, the mean (SD) age was 55.4 (16.3) years, 55.0% were women, and the prevalence of diabetes was 14.9%. These characteristics, as well as the distribution of index eGFR, were stable across the study period. The likelihood of receiving dialysis at eGFR levels of 10 to 24 mL/min/1.73 m2 generally increased over time. For example, the 1-year odds of initiating dialysis increased for every 3-year interval by 5.2% (adjusted odds ratio, 1.052; 95% CI, 1.004-1.102) among adults with an index eGFR of 20 to 24 mL/min/1.73 m2, by 6.6% (adjusted odds ratio, 1.066; 95% CI, 1.007-1.130) among adults with an eGFR of 16 to 17 mL/min/1.73 m2, and by 5.3% (adjusted odds ratio, 1.053; 95% CI, 1.008-1.100) among adults with an eGFR of 10 to 13 mL/min/1.73 m2, adjusting for age, sex, race, and diabetes. The incidence of new cases of ESKD was estimated to have potentially been 16% (95% CI, 13%-18%) lower if there were no changes in system-level practice patterns or other factors besides timing of initiating long-term dialysis from the initial 3-year interval (2001-2003) to the final interval (2016-2018) assessed in this study. CONCLUSIONS AND RELEVANCE The present results underscore the importance the timing of initiating long-term dialysis has on the size of the population of individuals with ESKD.
Collapse
Affiliation(s)
- Chi-Yuan Hsu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco.,Division of Research, Kaiser Permanente Northern California, Oakland
| | - Rishi V Parikh
- Division of Research, Kaiser Permanente Northern California, Oakland
| | - Leonid N Pravoverov
- Department of Nephrology, Kaiser Permanente Oakland Medical Center, Oakland, California
| | - Sijie Zheng
- Division of Research, Kaiser Permanente Northern California, Oakland.,Department of Nephrology, Kaiser Permanente Oakland Medical Center, Oakland, California.,Division of Medical Education, Department of Medicine, University of California, San Francisco, San Francisco
| | - David V Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco
| | - Thida C Tan
- Division of Research, Kaiser Permanente Northern California, Oakland
| | - Alan S Go
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco.,Division of Research, Kaiser Permanente Northern California, Oakland.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco.,Department of Medicine, Stanford University, Stanford, California.,Department of Health Research and Policy, Stanford University, Stanford, California
| |
Collapse
|
470
|
Golestaneh L, Cavanaugh KL, Lo Y, Karaboyas A, Melamed ML, Johns TS, Norris KC. Community Racial Composition and Hospitalization Among Patients Receiving In-Center Hemodialysis. Am J Kidney Dis 2020; 76:754-764. [PMID: 32673736 PMCID: PMC7844565 DOI: 10.1053/j.ajkd.2020.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
RATIONALE & OBJECTIVE Community racial composition has been shown to be associated with mortality in patients receiving maintenenance dialysis. It is unclear whether living in communities with predominantly Black residents is also associated with risk for hospitalization among patients receiving hemodialysis. STUDY DESIGN Retrospective analysis of prospectively collected data from a cohort of patients receiving hemodialysis. SETTING & PARTICIPANTS 4,567 patients treated in 154 dialysis facilities located in 127 unique zip codes and enrolled in US Dialysis Outcomes and Practice Patterns Study (DOPPS) phases 4 to 5 (2010-2015). EXPOSURE Tertile of percentage of Black residents within zip code of patients' dialysis facility, defined through a link to the American Community Survey. OUTCOME Rate of hospitalizations during the study period. ANALYTIC APPROACH Associations of patient-, facility-, and community-level variables with community's percentage of Black residents were assessed using analysis of variance, Kruskal-Wallis, or χ2/Fisher exact tests. Negative binomial regression was used to estimate the incidence rate ratio for hospitalizations between these communities, with and without adjustment for potential confounding variables. RESULTS Mean age of study patients was 62.7 years. 53% were White, 27% were Black, and 45% were women. Median and threshold percentages of Black residents in zip codes in which dialysis facilities were located were 34.2% and≥14.4% for tertile 3 and 1.0% and≤1.8% for tertile 1, respectively. Compared with those in tertile 1 facilities, patients in tertile 3 facilities were more likely to be younger, be Black, live in urban communities with lower socioeconomic status, have a catheter as vascular access, and have fewer comorbid conditions. Patients dialyzing in communities with the highest tertile of Black residents experienced a higher adjusted rate of hospitalization (adjusted incidence rate ratio, 1.32; 95% CI, 1.12-1.56) compared with those treated in communities within the lowest tertile. LIMITATIONS Potential residual confounding. CONCLUSIONS The risk for hospitalization for patients receiving maintenance dialysis is higher among those treated in communities with a higher percentage of Black residents after adjustment for dialysis care, patient demographics, and comorbid conditions. Understanding the cause of this association should be a priority of future investigation.
Collapse
Affiliation(s)
- Ladan Golestaneh
- Renal Division, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY.
| | - Kerri L Cavanaugh
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN
| | - Yungtai Lo
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | - Michal L Melamed
- Renal Division, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY
| | - Tanya S Johns
- Renal Division, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY
| | - Keith C Norris
- Division of General Internal Medicine, UCLA/David Geffen School of Medicine, Los Angeles, CA; Division of Nephrology, UCLA/David Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|
471
|
Elliott MJ, Love S, Donald M, Manns B, Donald T, Premji Z, Hemmelgarn BR, Grinman M, Lang E, Ronksley PE. Outpatient Interventions for Managing Acute Complications of Chronic Diseases: A Scoping Review and Implications for Patients With CKD. Am J Kidney Dis 2020; 76:794-805. [PMID: 32479925 DOI: 10.1053/j.ajkd.2020.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 04/02/2020] [Indexed: 11/11/2022]
Abstract
RATIONALE & OBJECTIVE Patients with chronic kidney disease (CKD) have high rates of emergency department (ED) use and hospitalization. Outpatient care may provide an alternative to ED and inpatient care in this population. We aimed to explore the scope of outpatient interventions used to manage acute complications of chronic diseases and highlight opportunities to adapt and test interventions in the CKD population. STUDY DESIGN Scoping review of quantitative and qualitative studies. SETTING & POPULATION Outpatient interventions for adults experiencing acute complications related to 1 of 5 eligible chronic diseases (ie, CKD, chronic respiratory disease, cardiovascular disease, cancer, and diabetes). SELECTION CRITERIA FOR STUDIES MEDLINE, EMBASE, CINAHL, Cochrane Central Register of Controlled Trials, grey literature, and conference abstracts were searched to December 2019. DATA EXTRACTION Intervention and study characteristics were extracted using standardized tools. ANALYTICAL APPROACH Quantitative data were summarized descriptively; qualitative data were summarized thematically. Our approach observed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) extension for scoping reviews. RESULTS 77 studies (25 randomized controlled trials, 29 observational, 12 uncontrolled before-after, 5 quasi-experimental, 4 qualitative, and 2 mixed method) describing 57 unique interventions were included. Of identified intervention types (hospital at home [n = 16], observation unit [n = 9], ED-based specialist service [n = 4], ambulatory program [n = 18], and telemonitoring [n = 10]), most were studied in chronic respiratory and cardiovascular disease populations. None targeted the CKD population. Interventions were delivered in the home, ED, hospital, and ambulatory setting by a variety of health care providers. Cost savings were demonstrated for most interventions, although improvements in other outcome domains were not consistently observed. LIMITATIONS Heterogeneity of included studies; lack of data for outpatient interventions for acute complications related to CKD. CONCLUSIONS Several interventions for outpatient management of acute complications of chronic disease were identified. Although none was specific to the CKD population, features could be adapted and tested to address the complex acute-care needs of patients with CKD.
Collapse
Affiliation(s)
- Meghan J Elliott
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada.
| | - Shannan Love
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Maoliosa Donald
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryn Manns
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Teagan Donald
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Zahra Premji
- Department of Libraries and Cultural Resources, University of Calgary, Calgary, AB, Canada
| | - Brenda R Hemmelgarn
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Michelle Grinman
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Eddy Lang
- Department of Emergency Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul E Ronksley
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
472
|
Gelfand SL, Mandel EI, Mendu ML, Lakin JR. Palliative Care in the Advancing American Kidney Health Initiative: A Call for Inclusion in Kidney Care Delivery Models. Am J Kidney Dis 2020; 76:877-882. [PMID: 33228851 PMCID: PMC9596188 DOI: 10.1053/j.ajkd.2020.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 07/15/2020] [Indexed: 11/21/2022]
Abstract
The Advancing American Kidney Health (AAKH) Initiative aims to promote high-value patient-centered care by improving access to and quality of treatment options for kidney failure. The 3 explicit goals of the initiative are to reduce the incidence of kidney failure, increase the number of available kidneys for transplantation, and increase transplantation and home dialysis. To ensure a patient-centered movement toward home dialysis modalities, actionable principles of palliative care, including systematic communication and customized treatment plans, should be incorporated into this policy. In this perspective, we describe 2 opportunities to strengthen the patience-centeredness of the AAKH Initiative through palliative care: (1) serious illness conversations should be required for all dialysis initiations in the End-Stage Renal Disease Treatment Choices model, and (2) conservative kidney management should be counted as a home modality alongside peritoneal dialysis and home hemodialysis. A serious illness conversation can help clinicians discern whether a patient’s goals and values are best respected by a home dialysis modality or whether a nondialytic strategy such as conservative kidney management should be considered. An intensive and careful patient- and family-centered selection process will be necessary to ensure that no patient is pressured to forego conventional dialysis.
Collapse
|
473
|
Groeneweg KE, Au YW, Duijs JMGJ, Florijn BW, van Kooten C, de Fijter JW, Reinders MEJ, van Zonneveld AJ, Bijkerk R. Diabetic nephropathy alters circulating long noncoding RNA levels that normalize following simultaneous pancreas-kidney transplantation. Am J Transplant 2020; 20:3451-3461. [PMID: 32353171 PMCID: PMC7754299 DOI: 10.1111/ajt.15961] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 01/25/2023]
Abstract
Simultaneous pancreas-kidney transplantation (SPKT) replaces kidney function and restores endogenous insulin secretion in patients with diabetic nephropathy (DN). Here, we aimed to identify circulating long noncoding RNAs (lncRNAs) that are associated with DN and vascular injury in the context of SPKT. Based on a pilot study and a literature-based selection of vascular injury-related lncRNAs, we assessed 9 candidate lncRNAs in plasma samples of patients with diabetes mellitus with a kidney function >35 mL/min/1.73 m2 (DM; n = 12), DN (n = 14), SPKT (n = 35), healthy controls (n = 15), and renal transplant recipients (KTx; n = 13). DN patients were also studied longitudinally before and 1, 6, and 12 months after SPKT. Of 9 selected lncRNAs, we found MALAT1, LIPCAR, and LNC-EPHA6 to be higher in DN compared with healthy controls. SPKT caused MALAT1, LIPCAR, and LNC-EPHA6 to normalize to levels of healthy controls, which was confirmed in the longitudinal study. In addition, we observed a strong association between MALAT1, LNC-EPHA6, and LIPCAR and vascular injury marker soluble thrombomodulin and a subset of angiogenic microRNAs (miR-27a, miR-130b, miR-152, and miR-340). We conclude that specific circulating lncRNAs associate with DN-related vascular injury and normalize after SPKT, suggesting that lncRNAs may provide a promising novel monitoring strategy for vascular integrity in the context of SPKT.
Collapse
Affiliation(s)
- Koen E. Groeneweg
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Yu Wah Au
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Jacques M. G. J. Duijs
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Barend W. Florijn
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Johan W. de Fijter
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Marlies E. J. Reinders
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology)Einthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
474
|
Daily inhalation of hydrogen gas has a blood pressure-lowering effect in a rat model of hypertension. Sci Rep 2020; 10:20173. [PMID: 33244027 PMCID: PMC7692487 DOI: 10.1038/s41598-020-77349-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/09/2020] [Indexed: 12/27/2022] Open
Abstract
A recent clinical study demonstrated that haemodialysis with a dialysate containing hydrogen (H2) improves blood pressure control in end-stage kidney disease. Herein, we examined whether H2 has a salutary effect on hypertension in animal models. We subjected 5/6 nephrectomised rats to inhalation of either H2 (1.3% H2 + 21% O2 + 77.7% N2) or control (21% O2 + 79% N2) gas mixture for 1 h per day. H2 significantly suppressed increases in blood pressure after 5/6 nephrectomy. The anti-hypertensive effect of H2 was also confirmed in rats in a stable hypertensive state 3 weeks after nephrectomy. To examine the detailed effects of H2 on hypertension, we used an implanted telemetry system to continuously monitor blood pressure. H2 exerted an anti-hypertensive effect not only during daytime rest, but also during night-time activities. Spectral analysis of blood pressure variability revealed that H2 improved autonomic imbalance, namely by suppressing the overly active sympathetic nervous system and augmenting parasympathetic nervous system activity; these effects co-occurred with the blood pressure-lowering effect. In conclusion, 1-h daily exposure to H2 exerts an anti-hypertensive effect in an animal model of hypertension.
Collapse
|
475
|
Loutradis C, Sarafidis PA, Ferro CJ, Zoccali C. Volume overload in hemodialysis: diagnosis, cardiovascular consequences, and management. Nephrol Dial Transplant 2020; 36:2182-2193. [PMID: 33184659 PMCID: PMC8643589 DOI: 10.1093/ndt/gfaa182] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Volume overload in haemodialysis (HD) patients associates with hypertension and cardiac dysfunction and is a major risk factor for all-cause and cardiovascular mortality in this population. The diagnosis of volume excess and estimation of dry weight is based largely on clinical criteria and has a notoriously poor diagnostic accuracy. The search for accurate and objective methods to evaluate dry weight and to diagnose subclinical volume overload has been intensively pursued over the last 3 decades. Most methods have not been tested in appropriate clinical trials and their usefulness in clinical practice remains uncertain, except for bioimpedance spectroscopy and lung ultrasound (US). Bioimpedance spectroscopy is possibly the most widely used method to subjectively quantify fluid distributions over body compartments and produces reliable and reproducible results. Lung US provides reliable estimates of extravascular water in the lung, a critical parameter of the central circulation that in large part reflects the left ventricular end-diastolic pressure. To maximize cardiovascular tolerance, fluid removal in volume-expanded HD patients should be gradual and distributed over a sufficiently long time window. This review summarizes current knowledge about the diagnosis, prognosis and treatment of volume overload in HD patients.
Collapse
Affiliation(s)
| | - Pantelis A Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charles J Ferro
- Department of Renal Medicine, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Carmine Zoccali
- CNR-IFC Clinical Epidemiology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| |
Collapse
|
476
|
Oetjens MT, Luo JZ, Chang A, Leader JB, Hartzel DN, Moore BS, Strande NT, Kirchner HL, Ledbetter DH, Justice AE, Carey DJ, Mirshahi T. Electronic health record analysis identifies kidney disease as the leading risk factor for hospitalization in confirmed COVID-19 patients. PLoS One 2020; 15:e0242182. [PMID: 33180868 PMCID: PMC7660530 DOI: 10.1371/journal.pone.0242182] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Empirical data on conditions that increase risk of coronavirus disease 2019 (COVID-19) progression are needed to identify high risk individuals. We performed a comprehensive quantitative assessment of pre-existing clinical phenotypes associated with COVID-19-related hospitalization. METHODS Phenome-wide association study (PheWAS) of SARS-CoV-2-positive patients from an integrated health system (Geisinger) with system-level outpatient/inpatient COVID-19 testing capacity and retrospective electronic health record (EHR) data to assess pre-COVID-19 pandemic clinical phenotypes associated with hospital admission (hospitalization). RESULTS Of 12,971 individuals tested for SARS-CoV-2 with sufficient pre-COVID-19 pandemic EHR data at Geisinger, 1604 were SARS-CoV-2 positive and 354 required hospitalization. We identified 21 clinical phenotypes in 5 disease categories meeting phenome-wide significance (P<1.60x10-4), including: six kidney phenotypes, e.g. end stage renal disease or stage 5 CKD (OR = 11.07, p = 1.96x10-8), six cardiovascular phenotypes, e.g. congestive heart failure (OR = 3.8, p = 3.24x10-5), five respiratory phenotypes, e.g. chronic airway obstruction (OR = 2.54, p = 3.71x10-5), and three metabolic phenotypes, e.g. type 2 diabetes (OR = 1.80, p = 7.51x10-5). Additional analyses defining CKD based on estimated glomerular filtration rate, confirmed high risk of hospitalization associated with pre-existing stage 4 CKD (OR 2.90, 95% CI: 1.47, 5.74), stage 5 CKD/dialysis (OR 8.83, 95% CI: 2.76, 28.27), and kidney transplant (OR 14.98, 95% CI: 2.77, 80.8) but not stage 3 CKD (OR 1.03, 95% CI: 0.71, 1.48). CONCLUSIONS This study provides quantitative estimates of the contribution of pre-existing clinical phenotypes to COVID-19 hospitalization and highlights kidney disorders as the strongest factors associated with hospitalization in an integrated US healthcare system.
Collapse
Affiliation(s)
| | | | | | | | | | - Bryn S. Moore
- Geisinger, Danville, Pennsylvania, United States of America
| | | | | | | | | | - David J. Carey
- Geisinger, Danville, Pennsylvania, United States of America
| | | |
Collapse
|
477
|
A propensity score-matched analysis indicates screening for asymptomatic coronary artery disease does not predict cardiac events in kidney transplant recipients. Kidney Int 2020; 99:431-442. [PMID: 33171171 DOI: 10.1016/j.kint.2020.10.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/09/2020] [Accepted: 10/02/2020] [Indexed: 12/14/2022]
Abstract
Screening for asymptomatic coronary artery disease prior to kidney transplantation aims to reduce peri- and post-operative cardiac events. It is uncertain if this is achieved. Here, we investigated whether pre-transplant screening with a stress test or coronary angiogram associated with any difference in major adverse cardiac events (MACE) up to five years post-transplantation. We examined a national prospective cohort recruited to the Access to Transplant and Transplant Outcome Measures study who received a kidney transplant between 2011-2017, and linked patient demographics and details of cardiac screening investigations to outcome data extracted from the Hospital Episode Statistics dataset and United Kingdom Renal Registry. Propensity score matched groups were analyzed using Kaplan-Meier and Cox survival analyses. Overall, 2572 individuals were transplanted in 18 centers; 51% underwent screening and the proportion undergoing screening by center ranged from 5-100%. The incidence of MACE at 90 days, one and five years was 0.9%, 2.1% and 9.4% respectively. After propensity score matching based on the presence or absence of screening, 1760 individuals were examined (880 each in screened and unscreened groups). There was no statistically significant association between screening and MACE at 90 days (hazard ratio 0.80, 95% Confidence Interval 0.31-2.05), one year (1.12, 0.51-2.47) or five years (1.31, 0.86-1.99). Age, male sex and history of ischemic heart disease were associated with MACE. Thus, there is no association between screening for asymptomatic coronary artery disease and MACE up to five years post-transplant. Practices involving unselected screening of transplant recipients should be reviewed.
Collapse
|
478
|
Chen J, Hamm LL, Bundy JD, Kumbala DR, Bodana S, Chandra S, Chen CS, Starcke CC, Guo Y, Schaefer CM, Lustigova E, Mahone E, Vadalia AM, Livingston T, Obst K, Hernandez J, Bokhari SR, Kleinpeter M, Alper AB, Lukitsch I, He H, Nieman DC, He J. Combination Treatment with Sodium Nitrite and Isoquercetin on Endothelial Dysfunction among Patients with CKD: A Randomized Phase 2 Pilot Trial. Clin J Am Soc Nephrol 2020; 15:1566-1575. [PMID: 33023894 PMCID: PMC7646238 DOI: 10.2215/cjn.02020220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 08/28/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Endothelial dysfunction is common among patients with CKD. We tested the efficacy and safety of combination treatment with sodium nitrite and isoquercetin on biomarkers of endothelial dysfunction in patients with CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This randomized, double-blind, placebo-controlled phase 2 pilot trial enrolled 70 patients with predialysis CKD. Thirty-five were randomly assigned to combination treatment with sodium nitrite (40 mg twice daily) and isoquercetin (225 mg once daily) for 12 weeks, and 35 were randomly assigned to placebo. The primary outcome was mean change in flow-mediated vasodilation over the 12-week intervention. Secondary and safety outcomes included biomarkers of endothelial dysfunction, inflammation, and oxidative stress as well as kidney function, methemoglobin, and adverse events. Intention-to-treat analysis was conducted. RESULTS Baseline characteristics, including age, sex, race, cigarette smoking, history of hypertension and diabetes, use of renin-angiotensin system blockers, BP, fasting glucose, lipid profile, kidney function, urine albumin-creatinine ratio, and endothelial biomarkers, were comparable between groups. Over the 12-week intervention, flow-mediated vasodilation increased 1.1% (95% confidence interval, -0.1 to 2.3) in the treatment group and 0.3% (95% confidence interval, -0.9 to 1.5) in the placebo group, and net change was 0.8% (95% confidence interval, -0.9 to 2.5). In addition, changes in biomarkers of endothelial dysfunction (vascular adhesion molecule-1, intercellular adhesion molecule-1, E-selectin, vWf, endostatin, and asymmetric dimethylarginine), inflammation (TNF-α, IL-6, C-reactive protein, IL-1 receptor antagonist, and monocyte chemoattractant protein-1), and oxidative stress (oxidized LDL and nitrotyrosines) were not significantly different between the two groups. Furthermore, changes in eGFR, urine albumin-creatinine ratio, methemoglobin, and adverse events were not significantly different between groups. CONCLUSIONS This randomized phase 2 pilot trial suggests that combination treatment with sodium nitrite and isoquercetin did not significantly improve flow-mediated vasodilation or other endothelial function biomarkers but also did not increase adverse events compared with placebo among patients with CKD. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Nitrite, Isoquercetin, and Endothelial Dysfunction (NICE), NCT02552888.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
- Department of Medicine, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana
| | - L. Lee Hamm
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Joshua D. Bundy
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | | | - Shirisha Bodana
- Department of Nephrology, Ochsner Health System, New Orleans, Louisiana
| | - Sehgal Chandra
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Chung-Shiuan Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Charlton C. Starcke
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Yajun Guo
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Caroline M. Schaefer
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Eva Lustigova
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Erin Mahone
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Aarti M. Vadalia
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Terra Livingston
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Katherine Obst
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - Jesus Hernandez
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Syed Rizwan Bokhari
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Myra Kleinpeter
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Arnold B. Alper
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ivo Lukitsch
- Department of Nephrology, Ochsner Health System, New Orleans, Louisiana
| | - Hua He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| | - David C. Nieman
- Human Performance Lab, Appalachian State University, Kannapolis, North Carolina
| | - Jiang He
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
- Tulane University Translational Science Institute, New Orleans, Louisiana
| |
Collapse
|
479
|
Peritubular Capillary Rarefaction: An Underappreciated Regulator of CKD Progression. Int J Mol Sci 2020; 21:ijms21218255. [PMID: 33158122 PMCID: PMC7662781 DOI: 10.3390/ijms21218255] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022] Open
Abstract
Peritubular capillary (PTC) rarefaction is commonly detected in chronic kidney disease (CKD) such as hypertensive nephrosclerosis and diabetic nephropathy. Moreover, PTC rarefaction prominently correlates with impaired kidney function and predicts the future development of end-stage renal disease in patients with CKD. However, it is still underappreciated that PTC rarefaction is a pivotal regulator of CKD progression, primarily because the molecular mechanisms of PTC rarefaction have not been well-elucidated. In addition to the established mechanisms (reduced proangiogenic factors and increased anti-angiogenic factors), recent studies discovered significant contribution of the following elements to PTC loss: (1) prompt susceptibility of PTC to injury, (2) impaired proliferation of PTC, (3) apoptosis/senescence of PTC, and (4) pericyte detachment from PTC. Mainly based on the recent and novel findings in basic research and clinical study, this review describes the roles of the above-mentioned elements in PTC loss and focuses on the major factors regulating PTC angiogenesis, the assessment of PTC rarefaction and its surrogate markers, and an overview of the possible therapeutic agents to mitigate PTC rarefaction during CKD progression. PTC rarefaction is not only a prominent histological characteristic of CKD but also a central driving force of CKD progression.
Collapse
|
480
|
Lin YT, Lo YC, Chiang HY, Jung CR, Wang CM, Chan TC, Kuo CC, Hwang BF. Particulate Air Pollution and Progression to Kidney Failure With Replacement Therapy: An Advanced CKD Registry–Based Cohort Study in Taiwan. Am J Kidney Dis 2020; 76:645-657.e1. [DOI: 10.1053/j.ajkd.2020.02.447] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 02/15/2020] [Indexed: 01/19/2023]
|
481
|
Zhai Y, Liu X, Yang Q, Dang X, Sun S, Shao X, Liu X, Wu Y, Bai H, Mao J, Dong Y, Ma Q, Kang G, Huang W, Zhu H, Fu R, Zhang A, Xu R, Sun Q, Jiang X, Lai L, Huang J, Luan J, Xia Z, Cui J, Zhao M, Wu X, Zhang Q, Li Y, Liu C, Wang M, Wang F, Tao Y, Huang Z, Zhang D, Zhao B, Chen C, Huang C, Gao X, Shen Q, Shen Y, Xu H. IPDN-China promotes the development of pediatric dialysis in China. Pediatr Nephrol 2020; 35:2163-2171. [PMID: 32529322 DOI: 10.1007/s00467-020-04630-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/16/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND In mainland China, dialysis for children with end-stage renal disease (ESRD) was not introduced until the 1980s. To describe the development of pediatric dialysis in different regions of China, a national pediatric dialysis network, namely, International Pediatric Dialysis Network-China (IPDN-China) ( www.pedpd.org.cn ), was launched in 2012. METHODS Original and updated information from the renal centers registered with the IPDN-China was collected between 2012 and 2016 from two sources, namely, the registry and the survey, and demographic features were analyzed. RESULTS Due to promotion by the IPDN-China, the number of registered renal centers increased from 12 to 39 between 2012 and 2016, with a significant increase in the coverage of the Chinese administrative divisions (from 26.5 to 67.6%) (p < 0.01); and the coverage of the pediatric (0~14 years old) population increased to nearly 90% in 2016. The distribution of renal centers indicated that East China had the highest average number of registered centers per million population (pmp) 0~14-year-old age group. Seventeen relatively large dialysis centers were distributed across 14 divisions. Various modalities of renal replacement therapy (RRT) were available in most centers. The IPDN-China has promoted collaborations between dieticians, psychologists, and social workers on dialysis teams to provide better service to children with ESRD and their families. The proportion of centers with all three types of paramedic support (i.e., dieticians, psychologists, and social workers) as well as the proportion of centers with a partial paramedic team significantly increased between 2012 (25.0%) and 2016 (69.2%) (p < 0.05). In terms of the point prevalent cases of patients (aged < 18 years), data from the survey of 39 registered centers revealed that the number of children with ESRD who were on RRT was 578 (49% received a kidney transplant) at the end of 2016, which was more than that reported in previous surveys. Data from the registry showed that 349 dialysis patients had been enrolled as of the end of 2016. The median age at RRT start was 9.5 years, and the leading cause of ESRD was congenital abnormalities of the kidney and urinary tract (CAKUT). CONCLUSIONS The IPDN-China has helped to promote the development of pediatric dialysis for ESRD in China by improving the organization of care for dialysis patients and increasing the availability and the quality of RRT for patients who need it. To improve knowledge about the epidemiology and outcomes of pediatric RRT around the country, a sustained effort needs to be made by the IPDN-China to increase the enrollment of dialysis patients and increase the number of registered centers in the future.
Collapse
Affiliation(s)
- Yihui Zhai
- Division of Nephrology, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, No. 399 Wanyuan Rd, Shanghai, 201102, People's Republic of China
| | - Xiaorong Liu
- Division of Nephrology, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Rd., West City District, Beijing, 100045, People's Republic of China
| | - Qing Yang
- Division of Nephrology, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiqiang Dang
- Division of Pediatric Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuzhen Sun
- Division of Pediatric Nephrology, Shandong Provincial Hospital, Jinan, China
| | - Xiaoshan Shao
- Division of Nephrology, Guiyang Children's Hospital, Guiyang, China
| | - Xuemei Liu
- Division of Nephrology, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Yubin Wu
- Division of Pediatric Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haitao Bai
- Division of Pediatric Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianhua Mao
- Division of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Dong
- Division of Nephrology, Anhui Provincial Children's Hospital, Hefei, China
| | - Qingshan Ma
- Division of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Guogui Kang
- Division of Pediatric Nephrology, Ningbo Women and Children's Hospital, Ningbo, China
| | - Wenyan Huang
- Division of Nephrology, Shanghai Children's Hospital, Shanghai, China
| | - Hongtao Zhu
- Division of Pediatric Nephrology, The First Hospital of Xinjiang Medical University, Urumqi, China
| | - Rui Fu
- Division of Nephrology, Jiangxi Children's Hospital, Nanchang, China
| | - Aihua Zhang
- Division of Nephrology, Nanjing Children's Hospital, Nanjing, China
| | - Ruiying Xu
- Division of Pediatric Nephrology, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Sun
- Division of Pediatric Nephrology, Qingdao Women and Children's Hospital, Qingdao, China
| | - Xiaoyun Jiang
- Division of Pediatric Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liwen Lai
- Division of Pediatric Nephrology, The third People's Hospital of Jingdezhen, Nanchang, China
| | - Jianping Huang
- Division of Nephrology, Bayi Children's Hospital, The Military General Hospital of Beijing PLA, Beijing, China
| | - Jiangwei Luan
- Division of Pediatric Nephrology, Wuhan Women and Children's Hospital, Wuhan, China
| | - Zhengkun Xia
- Division of Pediatric Nephrology, National Clinical Research Center of Kidney Disease, Jinlin Hospital, Nanjing, China
| | - Jianjun Cui
- Division of Nephrology, Shanxi Children's Hospital, Taiyuan, China
| | - Mengzhun Zhao
- Division of Pediatric Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiaolin Wu
- Division of Pediatric Nephrology, Hubei Women and Children's Hospital, Wuhan, China
| | - Qiuye Zhang
- Division of Pediatric Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yufeng Li
- Division of Pediatric Nephrology, Xin Hua Hospital Affiliated, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cuihua Liu
- Division of Nephrology, Henan Children's Hospital, Henan, China
| | - Mo Wang
- Division of Nephrology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Feiyan Wang
- Division of Nephrology, Urumqi Children's Hospital, Urumqi, China
| | - Yuhong Tao
- Division of Pediatric Nephrology, West China Second University Hospital, Chengdu, China
| | - Zhimin Huang
- Division of Pediatric Nephrology, The Affiliated Hospital of Guangdong University, Guangzhou, China
| | - Dongfeng Zhang
- Division of Nephrology, Children's Hospital of Hebei Province, Shijiazhuang, China
| | - Bo Zhao
- Division of Nephrology, Kunming Children's Hospital, Kunming, China
| | - Chaoying Chen
- Division of Nephrology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Chunlin Huang
- Division of Nephrology, The Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xia Gao
- Division of Pediatric Nephrology, Gansu Provincial Hospital, Lanzhou, China
| | - Qian Shen
- Division of Nephrology, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, No. 399 Wanyuan Rd, Shanghai, 201102, People's Republic of China.
| | - Ying Shen
- Division of Nephrology, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Rd., West City District, Beijing, 100045, People's Republic of China.
| | - Hong Xu
- Division of Nephrology, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, No. 399 Wanyuan Rd, Shanghai, 201102, People's Republic of China.
| | | |
Collapse
|
482
|
Park S, Kim M, Kim JE, Kim K, Park M, Kim YC, Joo KW, Kim YS, Lee H. Characteristics of kidney transplantation recipients over time in South Korea. Korean J Intern Med 2020; 35:1457-1467. [PMID: 32218102 PMCID: PMC7652657 DOI: 10.3904/kjim.2019.292] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/27/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND/AIMS Detailed nationwide information regarding the recent status and time trends of kidney transplantation (KT) in South Korea is limited. METHODS We performed a nationwide, population-based cohort study using the national claims database of Korea. We included KT recipients from 2008 to 2016, and their demographic and clinical characteristics were collected. The prognostic outcome was graft failure consisted of patient death and death-censored graft failure (DCGF). RESULTS We studied 14,601 KT recipients with median follow-up duration of 3.96 years. The median age at the time of transplantation consistently increased from the past, and proportion of underlying diabetes mellitus prominently increased, reaching 35.6% in 2016. The preemptive KT accounted for approximately 30% of the total transplantation cases. The recipients showed a 10-year cumulative graft survival rate of 71.8%, consisting of 10-year DCGF free survival of 77.6% and patient survival of 92.8%. Age ≥ 20 and < 30 years, age ≥ 70 years, underlying history of diabetes, non-preemptive transplantation, and poor compliance on tacrolimus and mycophenolic acid were the significant risk factors associated with worse DCGF outcome. The economic cost of KT showed prominently increasing trends, reaching a total insured fee of > 60,000,000$ in 2016. However, the expansion was mainly burdened by the national insurance service but not by the patients. CONCLUSION In South Korea, the number of kidney transplantation in elderly or in patients with comorbidities has been increasing. Complex clinical factors were associated with medication compliance and patient prognosis.
Collapse
Affiliation(s)
- Sehoon Park
- Department of Internal Medicine, Armed Forces Capital Hospital, Seongnam, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Myoungsuk Kim
- Department of Biomedical Research, Seoul National University Hospital, Seoul, Korea
| | - Ji Eun Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kwangsoo Kim
- Department of Biomedical Research, Seoul National University Hospital, Seoul, Korea
| | - Minsu Park
- Statistics and Data Center, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Correspondence to Hajeong Lee, M.D. Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea Tel: +82-2-2072-4905 Fax: +82-2-766-9662 E-mail:
| |
Collapse
|
483
|
Lu YA, Chen SW, Lee CC, Wu VCC, Fan PC, Kuo G, Chen JJ, Chu PH, Chang SH, Chang CH. Mid-term survival of patients with chronic kidney disease after extracorporeal membrane oxygenation. Interact Cardiovasc Thorac Surg 2020; 31:595-602. [PMID: 33005952 DOI: 10.1093/icvts/ivaa168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/30/2020] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Chronic kidney disease (CKD) impairs the elimination of fluids, electrolytes and metabolic wastes, which can affect the outcomes of extracorporeal membrane oxygenation (ECMO) treatment. This study aimed to elucidate the impact of CKD on in-hospital mortality and mid-term survival of adult patients who received ECMO treatment. METHODS Patients who received first-time ECMO treatment between 1 January 2003 and 31 December 2013 were included. Those with CKD were identified and matched to patients without CKD using a 1:2 ratio and were followed for 3 years. The study outcomes included in-hospital outcomes and the 3-year mortality rate. A subgroup analysis was conducted by comparing the dialytic patients with the non-dialytic CKD patients. RESULTS The study comprised 1008 CKD patients and 2016 non-CKD patients after propensity score matching. The CKD patients had higher in-hospital mortality rates [69.5% vs 62.2%; adjusted odds ratio 1.41; 95% confidence interval (CI) 1.15-1.72] than the non-CKD patients. The 3-year mortality rate was 80.4% in the CKD group and 68% in the non-CKD group (adjusted hazard ratio 1.17; 95% CI 1.06-1.28). The subgroup analysis showed that the 3-year mortality rates were 84.5% and 78.4% in the dialytic and non-dialytic patients, respectively. No difference in the 3-year mortality rate was noted between the 2 CKD subgroups (P = 0.111). CONCLUSIONS CKD was associated with increased risks of in-hospital and mid-term mortalities in patients who received ECMO treatment. Furthermore, no difference in survival was observed between the patients with end-stage renal disease and non-dialytic CKD patients.
Collapse
Affiliation(s)
- Yueh-An Lu
- Division of Nephrology, Kidney Research Center, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shao-Wei Chen
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Cardiothoracic and Vascular Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Cheng-Chia Lee
- Division of Nephrology, Kidney Research Center, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Victor Chien-Chia Wu
- Department of Cardiology, Chang Gung Memorial Hospital, Keelung Branch and Linkou Medical Center, Taoyuan City, Taiwan
| | - Pei-Chun Fan
- Division of Nephrology, Kidney Research Center, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - George Kuo
- Division of Nephrology, Kidney Research Center, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Jia-Jin Chen
- Division of Nephrology, Kidney Research Center, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Po-Hsien Chu
- Department of Cardiology, Chang Gung Memorial Hospital, Keelung Branch and Linkou Medical Center, Taoyuan City, Taiwan
| | - Shang-Hung Chang
- Department of Cardiology, Chang Gung Memorial Hospital, Keelung Branch and Linkou Medical Center, Taoyuan City, Taiwan
| | - Chih-Hsiang Chang
- Division of Nephrology, Kidney Research Center, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
484
|
Sheng K, Zhang P, Yao X, Li J, He Y, Chen J. Prognostic Machine Learning Models for First-Year Mortality in Incident Hemodialysis Patients: Development and Validation Study. JMIR Med Inform 2020; 8:e20578. [PMID: 33118948 PMCID: PMC7661257 DOI: 10.2196/20578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The first-year survival rate among patients undergoing hemodialysis remains poor. Current mortality risk scores for patients undergoing hemodialysis employ regression techniques and have limited applicability and robustness. OBJECTIVE We aimed to develop a machine learning model utilizing clinical factors to predict first-year mortality in patients undergoing hemodialysis that could assist physicians in classifying high-risk patients. METHODS Training and testing cohorts consisted of 5351 patients from a single center and 5828 patients from 97 renal centers undergoing hemodialysis (incident only). The outcome was all-cause mortality during the first year of dialysis. Extreme gradient boosting was used for algorithm training and validation. Two models were established based on the data obtained at dialysis initiation (model 1) and data 0-3 months after dialysis initiation (model 2), and 10-fold cross-validation was applied to each model. The area under the curve (AUC), sensitivity (recall), specificity, precision, balanced accuracy, and F1 score were used to assess the predictive ability of the models. RESULTS In the training and testing cohorts, 585 (10.93%) and 764 (13.11%) patients, respectively, died during the first-year follow-up. Of 42 candidate features, the 15 most important features were selected. The performance of model 1 (AUC 0.83, 95% CI 0.78-0.84) was similar to that of model 2 (AUC 0.85, 95% CI 0.81-0.86). CONCLUSIONS We developed and validated 2 machine learning models to predict first-year mortality in patients undergoing hemodialysis. Both models could be used to stratify high-risk patients at the early stages of dialysis.
Collapse
Affiliation(s)
- Kaixiang Sheng
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Zhang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiawei Li
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchun He
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
485
|
Murphy SL, Mahan JD, Troost JP, Srivastava T, Kogon AJ, Cai Y, Davis TK, Fernandez H, Fornoni A, Gbadegesin RA, Herreshoff E, Canetta PA, Nachman PH, Reeve BB, Selewski DT, Sethna CB, Wang CS, Bartosh SM, Gipson DS, Tuttle KR, CureGN Consortium 18GharaviAliAhnWooinAppelGerald B.AvasareRupali S.BabayevRevekkaBatalIbrahimBombackAndrew S.BrownEricCampenotEric S.CanettaPietroChanBrendaD’AgatiVivette D.FernandezHildaForoncewiczBartoszGhiggeriGian MarcoHinesWilliam H.JainNamrata G.KirylukKrzysztofLinFangmingLuganiFrancescaMarasaMaddalenaMarkowitzGlenMohanSumitMuchaKrzysztofNickolasThomas L.RadhakrishnanJaiRaoMaya K.Regunathan-ShenkRenuSanna-CherchiSimoneSantorielloDominickStokesMichael B.YuNatalieValeriAnthony M.ZvitiRonaldGreenbaumLarry A.SmoyerWilliam E.Al-UzriAmiraAshoorIsaAvilesDiegoBaraccoRossanaBarciaJohnBartoshSharonBelshaCraigBraunMichael C.ChishtiAftabClaesDonnaCramerCarlDavisKeefeErkanElifFeigDanielFreundlichMichaelHannaMelishaHidalgoGuillermoJainAmrishKhalidMydaKallashMahmoudMDLaneJerome C.MahanJohnMathewsNishaNesterCarlaPanCynthiaPatelHirenRevellAdelaideSreedharanRajasreeSteinkeJuliaWenderferScott E.WongCraig S.FalkRonaldCookWilliamDerebailVimalFogoAgnesGasimAdilGehrToddHarrisRaymondKiddJasonLaurinLouis-PhilippePendergraftWillPichetteVincentPowellThomas BrianRenfrowMatthew B.RoyalVirginieHolzmanLawrence B.AdlerSharonAlpersCharlesMatarRaed BouBrownElizabethChoiMichaelDellKatherine M.DukkipatiRamFervenzaFernando C.FornoniAlessiaGadegbekuCrystalGipsonPatrickHaselyLeahHingoraniSangeetaHladunewichMichelle A.HoganJonathanJeffersonJ. AshleyJhaveriKenarJohnstoneDuncan B.KaskelFrederickKoganAmyKoppJeffreyLemleyKevin V.DieguezLaura Malaga-MeyersKevinNeuAliciaO'ShaughnessyMichelle MarieO’TooleJohn F.ParekhRulanReichHeatherReidyKimberlyRondonHelbertSambandamKamalanathan K.SedorJohn R.SelewskiDavid T.SethnaChristine B.SchellingJeffreySperatiJohn C.Swiatecka-UrbanAgnesTrachtmanHowardTuttleKatherine R.WeisstuchJosephZhdanovaOlgaGillespieBrendaGipsonDebbie S.HerreshoffEmilyKretzlerMatthiasRobinsonBruce M.MarianiLauraTroostJonathan P.WladkowskiMatthewGuay-WoodfordLisa M.. Longitudinal Changes in Health-Related Quality of Life in Primary Glomerular Disease: Results From the CureGN Study. Kidney Int Rep 2020; 5:1679-1689. [PMID: 33102960 PMCID: PMC7569685 DOI: 10.1016/j.ekir.2020.06.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 04/27/2020] [Accepted: 06/09/2020] [Indexed: 11/18/2022] Open
Abstract
Introduction Prior cross-sectional studies suggest that health-related quality of life (HRQOL) worsens with more severe glomerular disease. This longitudinal analysis was conducted to assess changes in HRQOL with changing disease status. Methods Cure Glomerulonephropathy (CureGN) is a cohort of patients with minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, IgA vasculitis, or IgA nephropathy. HRQOL was assessed at enrollment and follow-up visits 1 to 3 times annually for up to 5 years with the Patient-Reported Outcomes Measurement Information System (PROMIS). Global health, anxiety, and fatigue domains were measured in all; mobility was measured in children; and sleep-related impairment was measured in adults. Linear mixed effects models were used to evaluate HRQOL responsiveness to changes in disease status. Results A total of 469 children and 1146 adults with PROMIS scores were included in the analysis. HRQOL improved over time in nearly all domains, though group-level changes were modest. Edema was most consistently associated with worse HRQOL across domains among children and adults. A greater number of symptoms also predicted worse HRQOL in all domains. Sex, age, obesity, and serum albumin were associated with some HRQOL domains. The estimated glomerular filtration rate (eGFR) was only associated with fatigue and adult physical health; proteinuria was not associated with any HRQOL domain in adjusted models. Conclusion HRQOL measures were responsive to changes in disease activity, as indicated by edema. HRQOL over time was not predicted by laboratory-based markers of disease. Patient-reported edema and number of symptoms were the strongest predictors of HRQOL, highlighting the importance of the patient experience in glomerular disease. HRQOL outcomes inform understanding of the patient experience for children and adults with glomerular diseases.
Collapse
Affiliation(s)
- Shannon L. Murphy
- Division of Nephrology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
- Correspondence: Shannon L. Murphy, University of North Carolina Kidney Center, 7024 Burnett-Womack / CB # 7155, Chapel Hill, North Carolina 27599, USA.
| | - John D. Mahan
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Jonathan P. Troost
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Tarak Srivastava
- Division of Nephrology, Department of Pediatrics, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Amy J. Kogon
- Division of Nephrology, Children’s Hospital of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yi Cai
- Division of Pediatric Nephrology, Helen DeVos Children’s Hospital, Grand Rapids, Michigan, USA
| | - T. Keefe Davis
- Division of Pediatric Nephrology, Washington University, St Louis, Missouri, USA
| | - Hilda Fernandez
- Division of Nephrology, Columbia University, New York, New York, USA
| | - Alessia Fornoni
- Department of Medicine, University of Miami, Miami, Florida, USA
| | | | - Emily Herreshoff
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pietro A. Canetta
- Division of Nephrology, Columbia University, New York, New York, USA
| | - Patrick H. Nachman
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bryce B. Reeve
- Department of Population Health Sciences, Center for Health Measurement, Duke University, Durham, North Carolina, USA
| | - David T. Selewski
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christine B. Sethna
- Division of Pediatric Nephrology, Cohen Children's Medical Center, New Hyde Park, New York, New York, USA
| | - Chia-shi Wang
- Division of Nephrology, Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Sharon M. Bartosh
- Division of Pediatric Nephrology, University of Wisconsin, Madison, Wisconsin, USA
| | - Debbie S. Gipson
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine R. Tuttle
- Division of Nephrology, Providence Health Care, University of Washington, Spokane, Washington, USA
| | | |
Collapse
|
486
|
Qian J, Lee T, Thamer M, Zhang Y, Crews DC, Allon M. Racial Disparities in the Arteriovenous Fistula Care Continuum in Hemodialysis Patients. Clin J Am Soc Nephrol 2020; 15:1796-1803. [PMID: 33082199 PMCID: PMC7769016 DOI: 10.2215/cjn.03600320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/17/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Arteriovenous fistulas are the optimal vascular access type for patients on hemodialysis. However, arteriovenous fistulas are used less frequently in Black than in White individuals. The arteriovenous fistula care continuum comprises a series of sequential steps. A better understanding is needed of where disparities exist along the continuum in order to mitigate racial differences in arteriovenous fistula use. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Using Medicare claims data from the United States Renal Data System, longitudinal analyses of patients ≥67 years initiating hemodialysis with a central venous catheter between July 1, 2010 and June 30, 2012 were performed. Three patient cohorts were identified: patients initiating hemodialysis with a catheter (n=41,814), patients with arteriovenous fistula placement within 6 months of dialysis initiation (n=14,077), and patients whose arteriovenous fistulas were successfully used within 6 months of placement (n=7068). Three arteriovenous fistula processes of care outcomes were compared between Blacks and Whites: (1) arteriovenous fistula creation, (2) successful arteriovenous fistula use, and (3) primary arteriovenous fistula patency after successful use. RESULTS An arteriovenous fistula was placed within 6 months of dialysis initiation in 37% of patients initiating dialysis with a catheter. Among the patients with arteriovenous fistula placement, the arteriovenous fistula was successfully used for dialysis within 6 months in 48% of patients. Among patients with successful arteriovenous fistula use, 21% maintained primary arteriovenous fistula patency at 3 years. After adjusting for competing risks, Black patients on hemodialysis were 10% less likely to undergo arteriovenous fistula placement (adjusted subdistribution hazard ratio, 0.90; 95% confidence interval, 0.87 to 0.94); 12% less likely to have successful arteriovenous fistula use after placement (adjusted subdistribution hazard ratio, 0.88; 95% confidence interval, 0.83 to 0.93); and 22% less likely to maintain primary arteriovenous fistula patency after successful use (subdistribution hazard ratio, 0.78; 95% confidence interval, 0.74 to 0.84). CONCLUSIONS Lower arteriovenous fistula use among Blacks older than 67 years of age treated with hemodialysis was attributable to each step along the continuum of arteriovenous fistula processes of care.
Collapse
Affiliation(s)
- Joyce Qian
- Medical Technology and Practice Patterns Institute, Bethesda, Maryland
| | - Timmy Lee
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama .,Veterans Affairs Medical Center, Medical Service and Section of Nephrology, Birmingham, Alabama
| | - Mae Thamer
- Medical Technology and Practice Patterns Institute, Bethesda, Maryland
| | - Yi Zhang
- Medical Technology and Practice Patterns Institute, Bethesda, Maryland
| | - Deidra C Crews
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Allon
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
487
|
Kitchlu A, Chan CT, Leung N, Chen S, Latcha S, Tam P. Perspectives From an Onconephrology Interest Group: Conference Report. Can J Kidney Health Dis 2020; 7:2054358120962589. [PMID: 33117547 PMCID: PMC7573731 DOI: 10.1177/2054358120962589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/16/2020] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION AND OBJECTIVE Onconephrology is a new and evolving field that deals with kidney complications in patients with cancer as well as the management of cancer in patients with preexisting kidney disease. With increasing numbers of patients with cancer with kidney-related complications, the field has garnered increased attention. Thus, an annual Greater Toronto Area Onconephrology Interest Group symposium was held in May 2019. The objective of the meeting was to demonstrate the junctures between oncology and nephrology by highlighting recent data regarding (1) kidney impairment in solid organ malignancies, (2) management and treatment of kidney cancer, (3) kidney impairment in hematologic malignancies, (4) malignancy and kidney transplantation, and (5) hyponatremia in patients with cancer. METHODS AND SOURCES OF INFORMATION Through a structured presentation, the group explored key topics discussed at a Kidney Disease Improving Global Outcomes (KDIGO) Controversies Conference on Onconephrology. Expert opinions, clinical trial findings, and publication summaries were used to illustrate patient and treatment-related considerations in onconephrology. KEY FINDINGS Kidney complications in patients with cancer are a central theme in onconephrology. An estimated 12% to 25% of patients with solid organ malignancies have chronic kidney disease (CKD), although in certain cancers, the prevalence of CKD is higher. Kidney impairment is also a common complication of some hematologic malignancies. The incidence of renal failure in patients with multiple myeloma is estimated at 18% to 56% and light chain cast nephropathy is seen in approximately 30% of these patients. In addition, there appears to be a bidirectional relationship between kidney cancer and CKD, with some data sets suggesting the risk increases as kidney function declines. Cancer is also of concern in patients with preexisting kidney disease. Kidney transplant recipients have a greater risk of cancer and a higher risk of cancer-related mortality. Kidney complications have also been associated with novel cancer therapies, such as immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy. An estimated 2% to 4% of patients initiating an immune checkpoint inhibitor may develop nephrotoxicity, whereas up to 40% of patients on CAR T-cell therapy experience cytokine release syndrome (CRS). Tumor lysis syndrome and electrolyte abnormalities, such as hyponatremia, have also been reported with CAR T-cell therapy. While the incidence and prevalence of hyponatremia vary depending on the cancer type and serum sodium cutoff point, hyponatremia may be seen in up to 46% of patients hospitalized in cancer centers. CONCLUSIONS Onconephrology is a developing field and the themes arising from this meeting indicate a need for greater collaboration between oncologists and nephrologists. Educational symposia and onconephrology fellowship programs may allow for improved cancer care for patients with kidney disease.
Collapse
Affiliation(s)
- Abhijat Kitchlu
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, ON, Canada
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Christopher T. Chan
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, ON, Canada
| | - Nelson Leung
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sheldon Chen
- Department of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Sheron Latcha
- Renal Division, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Tam
- Department of Medicine, Division of Nephrology, The Scarborough Hospital, Toronto, ON, Canada
| |
Collapse
|
488
|
Zheng Q, Yang H, Fu X, Huang Y, Wei R, Wang Y, Liu YN, Liu WJ. The efficacy and safety of roxadustat for anemia in patients with chronic kidney disease: a meta-analysis. Nephrol Dial Transplant 2020; 36:1603-1615. [PMID: 33051677 DOI: 10.1093/ndt/gfaa110] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is an increasing public health issue. Anemia, which is a complication of CKD, is associated with reduced quality of life and increased morbidity and mortality. Currently quite a few clinical studies have been conducted to compare roxadustat with epoetin alfa [all for dialysis-dependent (DD) patients] or placebo [all for nondialysis-dependent (NDD) patients]. Our meta-analysis aimed to investigate the efficacy and safety of roxadustat for anemia in patients with CKD. METHODS We thoroughly searched eight electronic resource databases for randomized controlled trials (RCTs) comparing the efficacy and safety between roxadustat versus epoetin alfa or placebo for the treatment of anemia in patients with CKD. RESULTS Four Phase 2 and two Phase 3 studies with 1010 participants were included. Hemoglobin (Hb) and transferrin levels were increased significantly in the roxadustat group versus those in the placebo {standard mean difference [SMD] 1.57 [95% confidence interval (CI) 1.17-1.98]; SMD 1.81 [95% CI 1.53-2.08]; respectively, both low-quality evidence} or epoetin alfa group [SMD 0.47 (95% CI 0.02-0.93), very low-quality evidence; SMD 1.05 (95% CI 0.81-1.29), low-quality evidence; respectively]. Hepcidin levels were reduced significantly in the roxadustat group versus those in the placebo [SMD -1.72 (95% CI -3.03 to -0.41), very low-quality evidence] or epoetin alfa group [SMD -0.23 (95% CI -0.43 to -0.02), low-quality evidence]. Ferritin and serum transferrin saturation (TSAT) levels were reduced significantly in the roxadustat group versus those in the placebo group [SMD -0.82 (95% CI -1.31 to -0.33); SMD -0.54 (95% CI -0.76 to -0.32), respectively; both low-quality evidence] and ferritin and TSAT levels in the roxadustat group were comparable to those in the epoetin alfa group [SMD 0.02 (95% CI -0.18-0.21); SMD 0.15 (95% CI -0.04-0.35), respectively, both low-quality evidence]. As for safety, the incidence of adverse events (AEs) in the roxadustat group was insignificantly different from that of the placebo group [risk ratio (RR) 0.99 (95% CI 0.83-1.18); P = 0.89, very low-quality evidence]. But the incidence of AEs in the roxadustat group was significantly higher than that in the epoetin alfa group [RR 1.25 (95% CI 1.01-1.54); P = 0.04, low-quality evidence]. There was no significant association between roxadustat and the incidence of serious AEs (SAEs) for both NDD and DD patients [RR 1.08 (95% CI 0.51-2.28) and RR 1.43 (95% CI 0.85-2.40), respectively, both very low-quality evidence]. CONCLUSION In this meta-analysis of RCTs, we found evidence that after the oral administration of roxadustat, NDD patients' Hb levels were increased effectively and DD patients' Hb levels were maintained effectively. The risk of SAEs was not observed with the short-term use of roxadustat. These findings support roxadustat for the treatment of anemia in patients with CKD.
Collapse
Affiliation(s)
- Qiyan Zheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Department of Endocrinology, Nephropathy of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Huisheng Yang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinwen Fu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Department of Endocrinology, Nephropathy of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yishan Huang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Department of Endocrinology, Nephropathy of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruojun Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China
| | - Yahui Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Department of Endocrinology, Nephropathy of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Ning Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Beijing, China.,Department of Endocrinology, Nephropathy of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Department of Endocrinology, Nephropathy of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
489
|
Sumida K, Dashputre AA, Potukuchi PK, Thomas F, Obi Y, Molnar MZ, Gatwood JD, Streja E, Kalantar-Zadeh K, Kovesdy CP. Laxative use in patients with advanced chronic kidney disease transitioning to dialysis. Nephrol Dial Transplant 2020; 36:2018-2026. [PMID: 33035325 DOI: 10.1093/ndt/gfaa205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Constipation is highly prevalent in patients with chronic kidney disease (CKD), particularly among those with end-stage renal disease (ESRD), partly due to their dietary restrictions, comorbidities and medications. Laxatives are typically used for constipation management; however, little is known about laxative use and its associated factors in patients with advanced CKD transitioning to ESRD. METHODS In a retrospective cohort of 102 477 US veterans transitioning to dialysis between October 2007 and March 2015, we examined the proportion of patients who filled a prescription for any type of laxative within each 6-month period over 36 months pre- and post-transition to ESRD. Factors associated with laxative use during the last 1-year pre-ESRD period were identified by multivariable logistic regression. RESULTS The proportion of patients prescribed laxatives increased as patients progressed to ESRD, peaking at 37.1% in the 6 months immediately following ESRD transition, then remaining fairly stable throughout the post-ESRD transition period. Among laxative users, stool softeners were the most commonly prescribed (∼30%), followed by hyperosmotics (∼20%), stimulants (∼10%), bulk formers (∼3%), chloride channel activator (<1%) and several combinations of these. The use of anticoagulants, oral iron supplements, non-opioid analgesics, antihistamines and opioid analgesics were among the factors independently associated with pre-ESRD laxative use. CONCLUSION The use of laxatives increased considerably as patients neared transition to ESRD, likely mirroring the increasing burden of drug-induced constipation during the ESRD transition period. Findings may provide novel insight into better management strategies to alleviate constipation symptoms and reduce medication requirements in patients with advanced CKD.
Collapse
Affiliation(s)
- Keiichi Sumida
- Department of Medicine, Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ankur A Dashputre
- Department of Medicine, Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA.,College of Graduate Health Sciences, Institute for Health Outcomes and Policy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Praveen K Potukuchi
- Department of Medicine, Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA.,College of Graduate Health Sciences, Institute for Health Outcomes and Policy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Fridtjof Thomas
- Department of Preventive Medicine, Division of Biostatistics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yoshitsugu Obi
- Department of Medicine, Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Miklos Z Molnar
- Department of Medicine, Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA.,James D. Eason Transplant Institute, Methodist University Hospital, Memphis, TN, USA.,Department of Surgery, Division of Transplant, University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Justin D Gatwood
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Nashville, TN, USA
| | - Elani Streja
- Division of Nephrology and Hypertension, Harold Simmons Center for Chronic Disease Research and Epidemiology, University of California-Irvine, Orange, CA, USA
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology and Hypertension, Harold Simmons Center for Chronic Disease Research and Epidemiology, University of California-Irvine, Orange, CA, USA
| | - Csaba P Kovesdy
- Department of Medicine, Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA.,Nephrology Section, Memphis VA Medical Center, Memphis, TN, USA
| |
Collapse
|
490
|
Apata IW, Kabbani S, Neu AM, Kear TM, D'Agata EMC, Levenson DJ, Kliger AS, Hicks LA, Patel PR. Opportunities to Improve Antibiotic Prescribing in Outpatient Hemodialysis Facilities: A Report From the American Society of Nephrology and Centers for Disease Control and Prevention Antibiotic Stewardship White Paper Writing Group. Am J Kidney Dis 2020; 77:757-768. [PMID: 33045256 PMCID: PMC7546947 DOI: 10.1053/j.ajkd.2020.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/02/2020] [Indexed: 11/11/2022]
Abstract
Antibiotic use is necessary in the outpatient hemodialysis setting because patients receiving hemodialysis are at increased risk for infections and sepsis. However, inappropriate antibiotic use can lead to adverse drug events, including adverse drug reactions and infections with Clostridioides difficile and antibiotic-resistant bacteria. Optimizing antibiotic use can decrease adverse events and improve infection cure rates and patient outcomes. The American Society of Nephrology and the US Centers for Disease Control and Prevention created the Antibiotic Stewardship in Hemodialysis White Paper Writing Group, comprising experts in antibiotic stewardship, infectious diseases, nephrology, and public health, to highlight strategies that can improve antibiotic prescribing for patients receiving maintenance hemodialysis. Based on existing evidence and the unique patient and clinical setting characteristics, the following strategies for improving antibiotic use are reviewed: expanding infection and sepsis prevention activities, standardizing blood culture collection processes, treating methicillin-susceptible Staphylococcus aureus infections with β-lactams, optimizing communication between nurses and prescribing providers, and improving data sharing across transitions of care. Collaboration among the Centers for Disease Control and Prevention; American Society of Nephrology; other professional societies such as infectious diseases, hospital medicine, and vascular surgery societies; and dialysis provider organizations can improve antibiotic use and the quality of care for patients receiving maintenance hemodialysis.
Collapse
Affiliation(s)
- Ibironke W Apata
- Centers for Disease Control and Prevention, Atlanta, MD; Division of Renal Medicine, Emory University School of Medicine, Atlanta, MD.
| | - Sarah Kabbani
- Centers for Disease Control and Prevention, Atlanta, MD
| | | | - Tamara M Kear
- M. Louise Fitzpatrick College of Nursing, Villanova University, Villanova, PA
| | | | | | | | - Lauri A Hicks
- Centers for Disease Control and Prevention, Atlanta, MD
| | - Priti R Patel
- Centers for Disease Control and Prevention, Atlanta, MD
| |
Collapse
|
491
|
Cocchi E, Nestor JG, Gharavi AG. Clinical Genetic Screening in Adult Patients with Kidney Disease. Clin J Am Soc Nephrol 2020; 15:1497-1510. [PMID: 32646915 PMCID: PMC7536756 DOI: 10.2215/cjn.15141219] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Expanded accessibility of genetic sequencing technologies, such as chromosomal microarray and massively parallel sequencing approaches, is changing the management of hereditary kidney diseases. Genetic causes account for a substantial proportion of pediatric kidney disease cases, and with increased utilization of diagnostic genetic testing in nephrology, they are now also detected at appreciable frequencies in adult populations. Establishing a molecular diagnosis can have many potential benefits for patient care, such as guiding treatment, familial testing, and providing deeper insights on the molecular pathogenesis of kidney diseases. Today, with wider clinical use of genetic testing as part of the diagnostic evaluation, nephrologists have the challenging task of selecting the most suitable genetic test for each patient, and then applying the results into the appropriate clinical contexts. This review is intended to familiarize nephrologists with the various technical, logistical, and ethical considerations accompanying the increasing utilization of genetic testing in nephrology care.
Collapse
Affiliation(s)
- Enrico Cocchi
- Division of Nephrology and Center for Precision Medicine and Genomics, Department of Medicine, Columbia University, New York, New York
- Department of Pediatrics, Universita' degli Studi di Torino, Torino, Italy
| | - Jordan Gabriela Nestor
- Division of Nephrology and Center for Precision Medicine and Genomics, Department of Medicine, Columbia University, New York, New York
| | - Ali G Gharavi
- Division of Nephrology and Center for Precision Medicine and Genomics, Department of Medicine, Columbia University, New York, New York
- Insititute of Genomic Medicine, Columbia University, New York, New York
| |
Collapse
|
492
|
Flythe JE, Hilliard TS, Ikeler K, Keller S, Gipson DS, Grandinetti AC, Nordyke RJ, Perrone RD, Roy-Chaudhury P, Unruh M, West M, Bocell F, Hurst FP. Toward Patient-Centered Innovation: A Conceptual Framework for Patient-Reported Outcome Measures for Transformative Kidney Replacement Devices. Clin J Am Soc Nephrol 2020; 15:1522-1530. [PMID: 32276947 PMCID: PMC7536748 DOI: 10.2215/cjn.00110120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Individuals with dialysis-dependent kidney failure experience considerable disease- and treatment-related decline in functional status and overall well-being. Despite these experiences, there have been few substantive technological advances in KRT in decades. As such, new federal initiatives seek to accelerate innovation. Historically, integration of patient perspectives into KRT product development has been limited. However, the US Food and Drug Administration recognizes the importance of incorporating patient perspectives into the total product life cycle (i.e., from product conception to postmarket surveillance) and encourages the consideration of patient-reported outcomes in regulatory-focused clinical trials when appropriate. Recognizing the significance of identifying patient-reported outcome measures (PROMs) that capture contemporary patient priorities, the Kidney Health Initiative, a public-private partnership between the American Society of Nephrology and US Food and Drug Administration, convened a workgroup to (1) develop a conceptual framework for a health-related quality of life PROM; (2) identify and map existing PROMs to the conceptual framework, prioritizing them on the basis of their supporting evidence for use in the regulatory environment; and (3) describe next steps for identifying PROMs for use in regulatory clinical trials of transformative KRT devices. This paper summarizes the proposed health-related quality-of-life PROM conceptual framework, maps and prioritizes PROMs, and identifies gaps and future needs to advance the development of rigorous, meaningful PROMS for use in clinical trials of transformative KRT devices.
Collapse
Affiliation(s)
- Jennifer E. Flythe
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina Kidney Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Cecil G. Sheps Center for Health Services Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Kourtney Ikeler
- American Institutes for Research, Chapel Hill, North Carolina
| | - San Keller
- American Institutes for Research, Chapel Hill, North Carolina
| | - Debbie S. Gipson
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | | | | | | | - Prabir Roy-Chaudhury
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina Kidney Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina
| | - Mark Unruh
- School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Melissa West
- Kidney Health Initiative and American Society of Nephrology, Washington, DC
| | - Fraser Bocell
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland
| | - Frank P. Hurst
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
493
|
Zhou H, Sim JJ, Shi J, Shaw SF, Lee MS, Neyer JR, Kovesdy CP, Kalantar-Zadeh K, Jacobsen SJ. β-Blocker Use and Risk of Mortality in Heart Failure Patients Initiating Maintenance Dialysis. Am J Kidney Dis 2020; 77:704-712. [PMID: 33010357 DOI: 10.1053/j.ajkd.2020.07.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/12/2020] [Indexed: 11/11/2022]
Abstract
RATIONAL & OBJECTIVE Beta-blockers are recommended for patients with heart failure (HF) but their benefit in the dialysis population is uncertain. Beta-blockers are heterogeneous, including with respect to their removal by hemodialysis. We sought to evaluate whether β-blocker use and their dialyzability characteristics were associated with early mortality among patients with chronic kidney disease with HF who transitioned to dialysis. STUDY DESIGN Retrospective cohort study. SETTING & PARTICIPANTS Adults patients with chronic kidney disease (aged≥18 years) and HF who initiated either hemodialysis or peritoneal dialysis during January 1, 2007, to June 30, 2016, within an integrated health system were included. EXPOSURES Patients were considered treated with β-blockers if they had a quantity of drug dispensed covering the dialysis transition date. OUTCOMES All-cause mortality within 6 months and 1 year or hospitalization within 6 months after transition to maintenance dialysis. ANALYTICAL APPROACH Inverse probability of treatment weights using propensity scores was used to balance covariates between treatment groups. Cox proportional hazard analysis and logistic regression were used to investigate the association between β-blocker use and study outcomes. RESULTS 3,503 patients were included in the study. There were 2,115 (60.4%) patients using β-blockers at transition. Compared with nonusers, the HR for all-cause mortality within 6 months was 0.79 (95% CI, 0.65-0.94) among users of any β-blocker and 0.68 (95% CI, 0.53-0.88) among users of metoprolol at transition. There were no observed differences in all-cause or cardiovascular-related hospitalization. LIMITATIONS The observational nature of our study could not fully account for residual confounding. CONCLUSIONS Beta-blockers were associated with a lower rate of mortality among incident hemodialysis patients with HF. Similar associations were not observed for hospitalizations within the first 6 months following transition to dialysis.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA.
| | - John J Sim
- Division of Nephrology and Hypertension, Kaiser Permanente Los Angeles Medical Center. Los Angeles, CA.
| | - Jiaxiao Shi
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Sally F Shaw
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Ming-Sum Lee
- Division of Cardiology, Kaiser Permanente Los Angeles Medical Center. Los Angeles, CA
| | - Jonathan R Neyer
- Division of Cardiology, Kaiser Permanente Los Angeles Medical Center. Los Angeles, CA
| | - Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology and Hypertension, University of California Irvine Medical Center, Irvine, CA
| | - Steven J Jacobsen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| |
Collapse
|
494
|
Levy DS, Grewal R, Le TH. Vitamin K deficiency: an emerging player in the pathogenesis of vascular calcification and an iatrogenic consequence of therapies in advanced renal disease. Am J Physiol Renal Physiol 2020; 319:F618-F623. [PMID: 32830534 DOI: 10.1152/ajprenal.00278.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Vascular calcification is a known complication of chronic kidney disease (CKD). The prevalence of vascular calcification in patients with non-dialysis-dependent CKD stages 3-5 has been shown to be as high as 79% (20). Vascular calcification has been associated with increased risk for mortality, hospital admissions, and cardiovascular disease (6, 20, 50, 55). Alterations in mineral and bone metabolism play a pivotal role in the pathogenesis of vascular calcification in CKD. As CKD progresses, levels of fibroblast growth factor-23, parathyroid hormone, and serum phosphorus increase and levels of 1,25-(OH)2 vitamin D decrease. These imbalances have been linked to the development of vascular calcification. More recently, additional factors have been found to play a role in vascular calcification. Matrix G1a protein (MGP) in its carboxylated form (cMGP) is a potent inhibitor of vascular calcification. Importantly, carboxylation of MGP is dependent on the cofactor vitamin K. In patients with CKD, vitamin K deficiency is prevalent and is exacerbated by warfarin, which is frequently used for anticoagulation. Insufficient bioavailability of vitamin K reduces the amount of cMGP available, and, therefore, it may lead to increased risk of vascular calcification. In vitro studies have shown that in the setting of a high-phosphate environment and vitamin K antagonism, human aortic valve interstitial cells become calcified. In this article, we discuss the pathophysiological consequence of vitamin K deficiency in the setting of altered mineral and bone metabolism, its prevalence, and clinical implications in patients with CKD.
Collapse
Affiliation(s)
- David S Levy
- University of Rochester School of Medicine, Rochester, New York
| | | | - Thu H Le
- University of Rochester School of Medicine, Rochester, New York
| |
Collapse
|
495
|
Ramadan OI, Naji A, Levine MH, Porrett PM, Dunn TB, Nazarian SM, Weinrieb RM, Kaminski M, Johnson D, Trofe-Clark J, Lorincz IS, Blumberg EA, Weikert BC, Bleicher M, Abt PL. Kidney transplantation and donation in the transgender population: A single-institution case series. Am J Transplant 2020; 20:2899-2904. [PMID: 32353210 DOI: 10.1111/ajt.15963] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/03/2020] [Accepted: 04/19/2020] [Indexed: 01/25/2023]
Abstract
The medical needs of the transgender population are increasingly recognized within the US health care system. Hormone therapy and gender-affirming surgery present distinct anatomic, hormonal, infectious, and psychosocial issues among transgender kidney transplant donors and recipients. We present the first reported experience with kidney transplantation and donation in transgender patients. A single-center case series (January 2014-December 2018) comprising 4 transgender kidney transplant recipients and 2 transgender living donors was constructed and analyzed. Experts in transplant surgery, transplant psychiatry, transplant infectious disease, pharmacy, and endocrinology were consulted to discuss aspects of care for these patients. Four transgender patients identified as male-to-female and 2 as female-to-male. Three of 6 had gender-affirming surgeries prior to transplant surgery, 1 of whom had further procedures posttransplant. Additionally, 4 patients were on hormone therapy. All 6 had psychiatric comorbidities. The 4 grafts have done well, with an average serum creatinine of 1.45 mg/dL at 2 years (range 1.01-1.85 mg/dL). However, patients encountered various postoperative complications, 1 of which was attributable to modified anatomy. Thus, transgender kidney transplant patients can present novel challenges in regard to surgical considerations as well as pre- and posttransplant care. Dedicated expertise is needed to optimize outcomes for this population.
Collapse
Affiliation(s)
- Omar I Ramadan
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ali Naji
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew H Levine
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paige M Porrett
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ty B Dunn
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susanna M Nazarian
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert M Weinrieb
- Department of Psychiatry, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mary Kaminski
- Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Johnson
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer Trofe-Clark
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Nephrology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ilona S Lorincz
- Division of Endocrinology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emily A Blumberg
- Division of Infectious Disease, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Blair C Weikert
- Division of Infectious Disease, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Melissa Bleicher
- Division of Nephrology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter L Abt
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
496
|
Abstract
The global burden of chronic kidney disease (CKD) is rapidly increasing with a projection of becoming the 5th most common cause of years of life lost globally by 2040. Aggravatingly, CKD is a major cause of catastrophic health expenditure. The costs of dialysis and transplantation consume up to 3% of the annual healthcare budget in high-income countries. Crucially, however, the onset and progression of CKD are often preventable. In 2020, the World Kidney Day campaign highlights the importance of preventive interventions-be it primary, secondary, or tertiary. This complementing article focuses on outlining and analyzing measures that can be implemented in every country to promote and advance CKD prevention. Primary prevention of kidney disease should focus on the modification of risk factors and addressing structural abnormalities of the kidney and urinary tracts, as well as exposure to environmental risk factors and nephrotoxins. In persons with pre-existing kidney disease, secondary prevention, including blood pressure optimization and glycemic control, should be the main goal of education and clinical interventions. In patients with advanced CKD, the management of comorbidities such as uremia and cardiovascular disease is a highly recommended preventative intervention to avoid or delay dialysis or kidney transplantation. Political efforts are needed to proliferate the preventive approach. While national policies and strategies for non-communicable diseases might be present in a country, specific policies directed toward education and awareness about CKD screening, management, and treatment are often lacking. Hence, there is an urgent need to increase awareness of the importance of preventive measures throughout populations, professionals, and policy makers.
Collapse
|
497
|
Umbro I, Fabiani V, Fabiani M, Angelico F, Del Ben M. A systematic review on the association between obstructive sleep apnea and chronic kidney disease. Sleep Med Rev 2020; 53:101337. [PMID: 32629235 DOI: 10.1016/j.smrv.2020.101337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 12/22/2022]
Abstract
This systematic review aims to provide a more comprehensive overview of the association between obstructive sleep apnea (OSA) and chronic kidney disease (CKD). A PubMed search was conducted using the terms "(obstructive sleep disorders OR obstructive sleep apnea OR obstructive sleep apnea syndrome) AND (kidney function OR renal outcome OR chronic kidney disease OR end stage renal disease)". Four hundred seventy four articles were initially retrieved, 227 in the last five years. We included articles with similar definitions of OSA, in particular only diagnosed by polysomnography, in order to exclude bias related to different diagnostic clinical tools, prediction algorithms and questionnaires that affected previous studies. Six cross-sectional and two retrospective studies were analyzed. There were a total of 8795 participants across all the studies with a mean age between 51 and 63 years. One study included only males but the proportion of males in the other studies ranged from 58 to 85.6%. The mean body mass index ranged from 25.2 to 32 kg/m2. The majority of studies considered indicate that OSA and CKD are significantly associated, in particular in their more severe categories. It is of great importance to set up a strong clinical collaboration between sleep medicine and nephrology.
Collapse
Affiliation(s)
- Ilaria Umbro
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy.
| | - Valerio Fabiani
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy
| | - Mario Fabiani
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Francesco Angelico
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Maria Del Ben
- Department of Clinical, Internal, Anesthetic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
498
|
Eckes T, Trautmann S, Djudjaj S, Beyer S, Patyna S, Schwalm S, Gauer S, Thomas D, Schaefer L, Boor P, Koch A, Pfeilschifter J. Consistent alteration of chain length-specific ceramides in human and mouse fibrotic kidneys. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158821. [PMID: 33010454 DOI: 10.1016/j.bbalip.2020.158821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Several studies revealed alterations of single sphingolipid species, such as chain length-specific ceramides, in plasma and serum of patients with kidney diseases. Here, we investigated whether such alterations occur in kidney tissue from patients and mice suffering from renal fibrosis, the common endpoint of chronic kidney diseases. METHODS Human fibrotic kidney samples were collected from nephrectomy specimens with hydronephrosis and/or pyelonephritis. Healthy parts from tumor nephrectomies served as nonfibrotic controls. Mouse fibrotic kidney samples were collected from male C57BL/6J mice treated with an adenine-rich diet for 14 days or were subjected to 7 days of unilateral ureteral obstruction (UUO). Kidneys of untreated mice and contralateral kidneys (UUO) served as respective controls. Sphingolipid levels were detected by LC-MS/MS. Fibrotic markers were analyzed by TaqMan® analysis and immunohistology. RESULTS Very long-chain ceramides Cer d18:1/24:0 and Cer d18:1/24:1 were significantly downregulated in both fibrotic human kidney cortex and fibrotic murine kidney compared to respective control samples. These effects correlate with upregulation of COL1α1, COL3α1 and αSMA expression in fibrotic human kidney cortex and fibrotic mouse kidney. CONCLUSION We have shown that very long-chain ceramides Cer d18:1/24:0 and Cer d18:1/24:1 are consistently downregulated in fibrotic kidney samples from human and mouse. Our findings support the use of in vivo murine models as appropriate translational means to understand the involvement of ceramides in human kidney diseases. In addition, our study raises interesting questions about the possible manipulation of ceramide metabolism to prevent progression of fibrosis and the use of ceramides as potential biomarkers of chronic kidney disease.
Collapse
Affiliation(s)
- Timon Eckes
- Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, Germany.
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Sonja Djudjaj
- Institute of Pathology, University Hospital of the RWTH Aachen, Germany
| | - Sandra Beyer
- Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Sammy Patyna
- Department of Nephrology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Stephanie Schwalm
- Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Stefan Gauer
- Department of Nephrology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Liliana Schaefer
- Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital of the RWTH Aachen, Germany; Division of Nephrology, University Hospital of the RWTH Aachen, Germany
| | - Alexander Koch
- Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, Germany
| |
Collapse
|
499
|
Rosner MH. Cancer Screening in Patients Undergoing Maintenance Dialysis: Who, What, and When. Am J Kidney Dis 2020; 76:558-566. [DOI: 10.1053/j.ajkd.2019.12.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/10/2019] [Indexed: 01/18/2023]
|
500
|
Siriwardana AN, Hoffman AT, Brennan FP, Li K, Brown MA. Impact of Renal Supportive Care on Symptom Burden in Dialysis Patients: A Prospective Observational Cohort Study. J Pain Symptom Manage 2020; 60:725-736. [PMID: 32389605 DOI: 10.1016/j.jpainsymman.2020.04.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/20/2020] [Accepted: 04/24/2020] [Indexed: 01/28/2023]
Abstract
CONTEXT Symptom burden is a strong predictor of reduced health-related quality of life and survival in patients with end-stage kidney disease. Renal supportive care (RSC) is a comprehensive approach shown to benefit symptoms in nondialysis conservatively managed patients, although its role in dialysis patients has not been reported. OBJECTIVES This study aimed to investigate the impacts of RSC intervention on symptoms in dialysis patients. METHODS Dialysis patients who were referred to an RSC clinic for symptom control between April 2010 and December 2017 were followed prospectively. Symptoms were scored using the Integrated Palliative care Outcomes Scale-Renal Inventory. Change in symptoms was analyzed at three visits and at final RSC visit within the study period. Correlation and linear regression were used to assess for effect modifiers. RESULTS A total of 127 dialysis patients attended the RSC clinic for symptom management. Median age was 74 years, 62% males, median dialysis vintage was 2.2 years, and median-modified Charlson Comorbidity Index was 7. Mean combined physical and emotional symptom score at baseline was 17.5 (SD 9.6), the most overwhelming/severe symptoms being difficulty sleeping (35%), pain (31%), lack of energy (31%), poor mobility (24%), and itch (22%). Eighty patients had follow-up to at least three RSC visits (median 3.1 months). There was significant improvement in combined physical and emotional symptom score during three clinic visits (18.1 vs. 14.2; mean change -3.8; 95% CI -5.7 to -1.9; P < 0.001), with greatest improvement in symptom scores for the five most severe symptoms (each P < 0.001). Follow-up of these 80 patients to final RSC visit (median 13.0 months) showed sustained reduction in mean combined physical and emotional symptom score (18.1 vs. 14.4; mean change -3.7; 95% CI -5.6 to -1.7; P < 0.001). These changes occurred without change in dialysis delivery. CONCLUSION RSC intervention that focuses on symptom control and patient-centered care is associated with improved total and individual symptom burden in dialysis patients. This supports a role for RSC as a management adjunct in these patients.
Collapse
Affiliation(s)
- Amanda N Siriwardana
- Department of Renal Medicine, St George Hospital, Sydney, New South Wales, Australia; Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.
| | - Anna T Hoffman
- Department of Renal Medicine, St George Hospital, Sydney, New South Wales, Australia
| | - Frank P Brennan
- Department of Renal Medicine, St George Hospital, Sydney, New South Wales, Australia
| | - Kelly Li
- Department of Renal Medicine, St George Hospital, Sydney, New South Wales, Australia
| | - Mark A Brown
- Department of Renal Medicine, St George Hospital, Sydney, New South Wales, Australia; University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|