301
|
Klimontov VV, Korbut AI. Albuminuric and non-albuminuric patterns of chronic kidney disease in type 2 diabetes. Diabetes Metab Syndr 2019; 13:474-479. [PMID: 30641747 DOI: 10.1016/j.dsx.2018.11.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/02/2018] [Indexed: 12/17/2022]
Abstract
A growing body of evidence supports a shift in the natural history of chronic kidney disease (CKD) in subjects with diabetes. Specifically, normoalbuminuric chronic kidney disease (NA-CKD), which is characterized by a decline in the glomerular filtration rate in the absence of a preceding or accompanying elevation of albuminuria, has become a widely prevalent variant of renal impairment in diabetes. Diabetic women and nonsmoking individuals with better glycemic control have a better chance of preserving normoalbuminuria, even in the case of declining renal function. The wide use of renin-angiotensin system blockers, advances in antihyperglycemic, antihypertensive, and hypolipidemic therapy, and smoking cessation are suspected to be responsible for an increasing proportion of NA-CKD among diabetic subjects with renal impairment. Significant differences in the sets of risk factors, renal morphology, comorbidity, and outcomes were found between the albuminuric and normoalbuminuric CKD patterns. NA-CKD, even if a more favorable option in terms of the risk of end-stage renal disease, is clearly associated with cardiovascular disease and its risk factors. The presence of NA-CKD in patients with diabetes increases the risk of myocardial infarction, stroke, and cardiovascular death. The study of the molecular pathways, clinical course, and outcomes of NA-CKD in diabetic subjects and the search for more specific diagnostic and treatment options are challenges for future research.
Collapse
Affiliation(s)
- Vadim V Klimontov
- Laboratory of Endocrinology, Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation.
| | - Anton I Korbut
- Laboratory of Endocrinology, Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| |
Collapse
|
302
|
Sagmeister MS, Taylor AE, Fenton A, Wall NA, Chanouzas D, Nightingale PG, Ferro CJ, Arlt W, Cockwell P, Hardy RS, Harper L. Glucocorticoid activation by 11β-hydroxysteroid dehydrogenase enzymes in relation to inflammation and glycaemic control in chronic kidney disease: A cross-sectional study. Clin Endocrinol (Oxf) 2019; 90:241-249. [PMID: 30358903 PMCID: PMC6334281 DOI: 10.1111/cen.13889] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Patients with chronic kidney disease (CKD) have dysregulated cortisol metabolism secondary to changes in 11β-hydroxysteroid dehydrogenase (11β-HSD) enzymes. The determinants of this and its clinical implications are poorly defined. METHODS We performed a cross-sectional study to characterize shifts in cortisol metabolism in relation to renal function, inflammation and glycaemic control. Systemic activation of cortisol by 11β-HSD was measured as the metabolite ratio (tetrahydrocortisol [THF]+5α-tetrahydrocortisol [5αTHF])/tetrahydrocortisone (THE) in urine. RESULTS The cohort included 342 participants with a median age of 63 years, median estimated glomerular filtration rate (eGFR) of 28 mL/min/1.73 m2 and median urine albumin-creatinine ratio of 35.5 mg/mmol. (THF+5αTHF)/THE correlated negatively with eGFR (Spearman's ρ = -0.116, P = 0.032) and positively with C-reactive protein (ρ = 0.208, P < 0.001). In multivariable analysis, C-reactive protein remained a significant independent predictor of (THF+5αTHF)/THE, but eGFR did not. Elevated (THF+5αTHF)/THE was associated with HbA1c (ρ = 0.144, P = 0.008) and diabetes mellitus (odds ratio for high vs low tertile of (THF+5αTHF)/THE 2.57, 95% confidence interval 1.47-4.47). Associations with diabetes mellitus and with HbA1c among the diabetic subgroup were independent of eGFR, C-reactive protein, age, sex and ethnicity. CONCLUSIONS In summary, glucocorticoid activation by 11β-HSD in our cohort comprising a spectrum of renal function was associated with inflammation and impaired glucose control.
Collapse
Affiliation(s)
- Michael S. Sagmeister
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Angela E. Taylor
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
| | - Anthony Fenton
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Nadezhda A. Wall
- Institute of Clinical SciencesUniversity of BirminghamBirminghamUK
| | - Dimitrios Chanouzas
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Peter G. Nightingale
- Institute of Translational MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Charles J. Ferro
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Wiebke Arlt
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
| | - Paul Cockwell
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Rowan S. Hardy
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
- Institute of Inflammation and Ageing, ARUK Rheumatoid Arthritis Centre of Excellence, and MRC ARUK Centre for Musculoskeletal AgeingUniversity of BirminghamBirminghamUK
| | - Lorraine Harper
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
- Institute of Clinical SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
303
|
Amdur RL, Feldman HI, Dominic EA, Anderson AH, Beddhu S, Rahman M, Wolf M, Reilly M, Ojo A, Townsend RR, Go AS, He J, Xie D, Thompson S, Budoff M, Kasner S, Kimmel PL, Kusek JW, Raj DS. Use of Measures of Inflammation and Kidney Function for Prediction of Atherosclerotic Vascular Disease Events and Death in Patients With CKD: Findings From the CRIC Study. Am J Kidney Dis 2018; 73:344-353. [PMID: 30545708 DOI: 10.1053/j.ajkd.2018.09.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Abstract
RATIONALE & OBJECTIVE Traditional risk estimates for atherosclerotic vascular disease (ASVD) and death may not perform optimally in the setting of chronic kidney disease (CKD). We sought to determine whether the addition of measures of inflammation and kidney function to traditional estimation tools improves prediction of these events in a diverse cohort of patients with CKD. STUDY DESIGN Observational cohort study. SETTING & PARTICIPANTS 2,399 Chronic Renal Insufficiency Cohort (CRIC) Study participants without a history of cardiovascular disease at study entry. PREDICTORS Baseline plasma levels of biomarkers of inflammation (interleukin 1β [IL-1β], IL-1 receptor antagonist, IL-6, tumor necrosis factor α [TNF-α], transforming growth factor β, high-sensitivity C-reactive protein, fibrinogen, and serum albumin), measures of kidney function (estimated glomerular filtration rate [eGFR] and albuminuria), and the Pooled Cohort Equation probability (PCEP) estimate. OUTCOMES Composite of ASVD events (incident myocardial infarction, peripheral arterial disease, and stroke) and death. ANALYTICAL APPROACH Cox proportional hazard models adjusted for PCEP estimates, albuminuria, and eGFR. RESULTS During a median follow-up of 7.3 years, 86, 61, 48, and 323 participants experienced myocardial infarction, peripheral arterial disease, stroke, or death, respectively. The 1-decile greater levels of IL-6 (adjusted HR [aHR], 1.12; 95% CI, 1.08-1.16; P<0.001), TNF-α (aHR, 1.09; 95% CI, 1.05-1.13; P<0.001), fibrinogen (aHR, 1.07; 95% CI, 1.03-1.11; P<0.001), and serum albumin (aHR, 0.96; 95% CI, 0.93-0.99; P<0.002) were independently associated with the composite ASVD-death outcome. A composite inflammation score (CIS) incorporating these 4 biomarkers was associated with a graded increase in risk for the composite outcome. The incidence of ASVD-death increased across the quintiles of risk derived from PCEP, kidney function, and CIS. The addition of eGFR, albuminuria, and CIS to PCEP improved (P=0.003) the area under the receiver operating characteristic curve for the composite outcome from 0.68 (95% CI, 0.66-0.71) to 0.73 (95% CI, 0.71-0.76). LIMITATIONS Data for cardiovascular death were not available. CONCLUSIONS Biomarkers of inflammation and measures of kidney function are independently associated with incident ASVD events and death in patients with CKD. Traditional cardiovascular risk estimates could be improved by adding markers of inflammation and measures of kidney function.
Collapse
Affiliation(s)
- Richard L Amdur
- Department of Surgery, George Washington University, Washington, DC
| | - Harold I Feldman
- Renal Electrolyte and Hypertension Division, University of Pennsylvania, PA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, PA
| | | | - Amanda H Anderson
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, PA
| | - Srinivasan Beddhu
- Division of Nephrology, University of Utah School of Medicine, Salt Lake City, UT
| | - Mahboob Rahman
- Division of Nephrology and Hypertension, Case Western Reserve University, OH
| | - Myles Wolf
- Division of Nephrology, Duke University, Durham, NC
| | - Muredach Reilly
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research, Columbia University College of Physician and Surgeon, New York, NY
| | - Akinlolu Ojo
- University of Arizona School of Medicine, Tucson, AZ
| | - Raymond R Townsend
- Renal Electrolyte and Hypertension Division, University of Pennsylvania, PA
| | - Alan S Go
- Kaiser Permanente Division of Research, Oakland, CA
| | - Jiang He
- Department of Epidemiology, Tulane University, LA
| | - Dawei Xie
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, PA
| | - Sally Thompson
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, PA
| | - Matthew Budoff
- Division of Cardiology, Los Angeles Biomedical Research Institute at Harbor-UCLA, Torrance, CA
| | - Scott Kasner
- Division of Vascular Neurology, University of Pennsylvania, PA
| | - Paul L Kimmel
- Division of Kidney Urologic and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - John W Kusek
- Division of Kidney Urologic and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University, Washington, DC.
| | | |
Collapse
|
304
|
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) is associated with bone loss and fractures. The purpose of this review is to provide clinicians with an overview of the underlying pathogenesis of CKD-associated osteoporosis, and a summary of the current diagnostic and therapeutic approaches to this disease. RECENT FINDINGS In 2017, the Kidney Disease Improving Global Outcomes Committee on Bone Quality updated their guidelines to include screening for osteoporosis and fracture risk by dual energy X-ray absorptiometry in patients with CKD. Once a diagnosis of osteoporosis and/or fracture risk is established, it is not clear how nephrologists should manage their patients. Patients with CKD should be screened for CKD-associated osteoporosis and considered for strategies that prevent bone loss and fractures. Assessment of bone turnover via imaging, biochemical testing, or bone biopsy can help guide the choice of therapy. Randomized controlled trials are needed to assess safety and efficacy of treatments to prevent bone loss and fractures.
Collapse
Affiliation(s)
- Pascale Khairallah
- Division of Nephrology, Department of Medicine, Columbia University Medical Center, 622 West 168th Street, PH2-124, New York City, NY, 10032, USA
| | - Thomas L Nickolas
- Division of Nephrology, Department of Medicine, Columbia University Medical Center, 622 West 168th Street, PH2-124, New York City, NY, 10032, USA.
| |
Collapse
|
305
|
Lodebo BT, Shah A, Kopple JD. Is it Important to Prevent and Treat Protein-Energy Wasting in Chronic Kidney Disease and Chronic Dialysis Patients? J Ren Nutr 2018; 28:369-379. [DOI: 10.1053/j.jrn.2018.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/30/2018] [Accepted: 04/05/2018] [Indexed: 01/05/2023] Open
|
306
|
da Silva EN, Baker A, Alshekaili J, Karpe K, Cook MC. A randomized trial of serological and cellular responses to hepatitis B vaccination in chronic kidney disease. PLoS One 2018; 13:e0204477. [PMID: 30303980 PMCID: PMC6179249 DOI: 10.1371/journal.pone.0204477] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 09/07/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with an increased risk of hepatitis B infection and impaired seroconversion to hepatitis B vaccine (HBV). Studies examining augmented vaccine schedules to enhance seroconversion have so far been inconclusive. Furthermore, the defects responsible for impaired vaccine immunity in CKD have not yet been identified. METHODS We studied serological and cellular responses to HBV in CKD to identify a defect in vaccine-induced cellular responses that could account for impaired seroconversion in CKD and clarify the effects of an augmented vaccine dose schedule. We compared these results with responses to seasonal influenza vaccination (Fluvax). RESULTS We found a clear benefit in rates and magnitude of seroconversion after an augmented 40mcg HBV dose schedule in CKD. This permitted comparison of responders and non-responders. Serological non-responders with CKD exhibited reduction in CXCR3+CCR6- CXCR5+ memory T cells at baseline. Unlike Fluvax, HBV elicited a poor plasmablast (PB) response. Both vaccinations induced activation of the CXCR3+CCR6- CCR7- subset of circulating T follicular helper cells (cTFH), although this response was impaired in CKD after HBV. CONCLUSIONS CKD confers a specific T cell defect that contributes to the impaired seroconversion to HBV.
Collapse
Affiliation(s)
- Elizabeth N da Silva
- Division of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Translational Research Unit, Canberra Hospital, Garran, ACT, Australia.,Department of Immunology, Canberra Hospital, Garran, ACT, Australia
| | - Alan Baker
- Department of Immunology, Liverpool Hospital, Liverpool, NSW, Australia
| | - Jalila Alshekaili
- Division of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Translational Research Unit, Canberra Hospital, Garran, ACT, Australia
| | - Krishna Karpe
- Department of Renal Medicine, The Canberra Hospital, Garran, ACT, Australia
| | - Matthew C Cook
- Division of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Translational Research Unit, Canberra Hospital, Garran, ACT, Australia.,Department of Immunology, Canberra Hospital, Garran, ACT, Australia
| |
Collapse
|
307
|
Abramowitz MK, Paredes W, Zhang K, Brightwell CR, Newsom JN, Kwon HJ, Custodio M, Buttar RS, Farooq H, Zaidi B, Pai R, Pessin JE, Hawkins M, Fry CS. Skeletal muscle fibrosis is associated with decreased muscle inflammation and weakness in patients with chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1658-F1669. [PMID: 30280599 DOI: 10.1152/ajprenal.00314.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Muscle dysfunction is an important cause of morbidity among patients with chronic kidney disease (CKD). Although muscle fibrosis is present in a CKD rodent model, its existence in humans and its impact on physical function are currently unknown. We examined isometric leg extension strength and measures of skeletal muscle fibrosis and inflammation in vastus lateralis muscle from CKD patients ( n = 10) and healthy, sedentary controls ( n = 10). Histochemistry and immunohistochemistry were used to assess muscle collagen and macrophage and fibro/adipogenic progenitor (FAP) cell populations, and RT-qPCR was used to assess muscle-specific inflammatory marker expression. Muscle collagen content was significantly greater in CKD compared with control (18.8 ± 2.1 vs. 11.7 ± 0.7% collagen area, P = 0.008), as was staining for collagen I, pro-collagen I, and a novel collagen-hybridizing peptide that binds remodeling collagen. Muscle collagen was inversely associated with leg extension strength in CKD ( r = -0.74, P = 0.01). FAP abundance was increased in CKD, was highly correlated with muscle collagen ( r = 0.84, P < 0.001), and was inversely associated with TNF-α expression ( r = -0.65, P = 0.003). TNF-α, CD68, CCL2, and CCL5 mRNA were significantly lower in CKD than control, despite higher serum TNF-α and IL-6. Immunohistochemistry confirmed fewer CD68+ and CD11b+ macrophages in CKD muscle. In conclusion, skeletal muscle collagen content is increased in humans with CKD and is associated with functional parameters. Muscle fibrosis correlated with increased FAP abundance, which may be due to insufficient macrophage-mediated TNF-α secretion. These data provide a foundation for future research elucidating the mechanisms responsible for this newly identified human muscle pathology.
Collapse
Affiliation(s)
| | - William Paredes
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Kehao Zhang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Camille R Brightwell
- Department of Nutrition and Metabolism, University of Texas Medical Branch , Galveston, Texas
| | - Julia N Newsom
- Department of Nutrition and Metabolism, University of Texas Medical Branch , Galveston, Texas
| | - Hyok-Joon Kwon
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers-The State University of New Jersey , New Brunswick, New Jersey
| | - Matthew Custodio
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Rupinder S Buttar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Hina Farooq
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Bushra Zaidi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Rima Pai
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Meredith Hawkins
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch , Galveston, Texas
| |
Collapse
|
308
|
Smykiewicz P, Segiet A, Keag M, Żera T. Proinflammatory cytokines and ageing of the cardiovascular-renal system. Mech Ageing Dev 2018; 175:35-45. [DOI: 10.1016/j.mad.2018.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/01/2018] [Accepted: 07/19/2018] [Indexed: 12/11/2022]
|
309
|
Inflammation-Related Mechanisms in Chronic Kidney Disease Prediction, Progression, and Outcome. J Immunol Res 2018; 2018:2180373. [PMID: 30271792 PMCID: PMC6146775 DOI: 10.1155/2018/2180373] [Citation(s) in RCA: 382] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Persistent, low-grade inflammation is now considered a hallmark feature of chronic kidney disease (CKD), being involved in the development of all-cause mortality of these patients. Although substantial improvements have been made in clinical care, CKD remains a major public health burden, affecting 10–15% of the population, and its prevalence is constantly growing. Due to its insidious nature, CKD is rarely diagnosed in early stages, and once developed, its progression is unfortunately irreversible. There are many factors that contribute to the setting of the inflammatory status in CKD, including increased production of proinflammatory cytokines, oxidative stress and acidosis, chronic and recurrent infections, altered metabolism of adipose tissue, and last but not least, gut microbiota dysbiosis, an underestimated source of microinflammation. In this scenario, a huge step forward was made by the increasing progression of omics approaches, specially designed for identification of biomarkers useful for early diagnostic and follow-up. Recent omics advances could provide novel insights in deciphering the disease pathophysiology; thus, identification of circulating biomarker panels using state-of-the-art proteomic technologies could improve CKD early diagnosis, monitoring, and prognostics. This review aims to summarize the recent knowledge regarding the relationship between inflammation and CKD, highlighting the current proteomic approaches, as well as the inflammasomes and gut microbiota dysbiosis involvement in the setting of CKD, culminating with the troubling bidirectional connection between CKD and renal malignancy, raised on the background of an inflammatory condition.
Collapse
|
310
|
Ishigami J, Matsushita K. Clinical epidemiology of infectious disease among patients with chronic kidney disease. Clin Exp Nephrol 2018; 23:437-447. [PMID: 30178234 PMCID: PMC6435626 DOI: 10.1007/s10157-018-1641-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022]
Abstract
Infectious disease is recognized as an important complication among patients with end-stage renal disease, contributing to excess morbidity and health care costs. However, recent epidemiological studies have revealed that even mild to moderate stages of chronic kidney disease (CKD) substantially increase risk of infection. Regarding underlying mechanisms, evidence suggests various aspects of altered immune response in patients with CKD including impaired function of T cells, B cells and neutrophil. Multiple conditions surrounding CKD, such as older age, diabetes, and cardiovascular disease are important contributors in the increased susceptibility to infection in this population. In addition, several mechanisms impairing immune function have been hypothesized including accumulated uremic toxins, increased oxidative stress, endothelial dysfunction, low-grade inflammation, and mineral and bone disorders. In terms of prevention strategies, influenza and pneumococcal vaccines are most feasible and important. Nevertheless, the extent of vaccine utilization in CKD has not been well documented. In addition, antibody response to vaccination may be reduced in CKD patients, and thus a vaccine delivery strategy (e.g., dose and frequency) may need to be optimized among patients with CKD. Through this review, we demonstrate that infection is a major but underrecognized complication of CKD. As CKD is recognized as a serious public health issue, dedicated research is needed to better characterize the burden of infectious disease associated with CKD, understand the pathophysiology of infection in patients with CKD, and develop effective strategies to prevent infection and its sequela in this high risk population.
Collapse
Affiliation(s)
- Junichi Ishigami
- Department of Epidemiology, Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, 2024 E. Monument St., Suite 2-600, Baltimore, MD, 21287, USA.
| | - Kunihiro Matsushita
- Department of Epidemiology, Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, 2024 E. Monument St., Suite 2-600, Baltimore, MD, 21287, USA
| |
Collapse
|
311
|
Abstract
Most older individuals develop inflammageing, a condition characterized by elevated levels of blood inflammatory markers that carries high susceptibility to chronic morbidity, disability, frailty, and premature death. Potential mechanisms of inflammageing include genetic susceptibility, central obesity, increased gut permeability, changes to microbiota composition, cellular senescence, NLRP3 inflammasome activation, oxidative stress caused by dysfunctional mitochondria, immune cell dysregulation, and chronic infections. Inflammageing is a risk factor for cardiovascular diseases (CVDs), and clinical trials suggest that this association is causal. Inflammageing is also a risk factor for chronic kidney disease, diabetes mellitus, cancer, depression, dementia, and sarcopenia, but whether modulating inflammation beneficially affects the clinical course of non-CVD health problems is controversial. This uncertainty is an important issue to address because older patients with CVD are often affected by multimorbidity and frailty - which affect clinical manifestations, prognosis, and response to treatment - and are associated with inflammation by mechanisms similar to those in CVD. The hypothesis that inflammation affects CVD, multimorbidity, and frailty by inhibiting growth factors, increasing catabolism, and interfering with homeostatic signalling is supported by mechanistic studies but requires confirmation in humans. Whether early modulation of inflammageing prevents or delays the onset of cardiovascular frailty should be tested in clinical trials.
Collapse
Affiliation(s)
- Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA.
| | - Elisa Fabbri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
312
|
Gunes H, Sokmen A, Kaya H, Gungor O, Kerkutluoglu M, Guzel FB, Sokmen G. Evaluation of Atrial Electromechanical Delay to Predict Atrial Fibrillation in Hemodialysis Patients. ACTA ACUST UNITED AC 2018; 54:medicina54040058. [PMID: 30344289 PMCID: PMC6174336 DOI: 10.3390/medicina54040058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022]
Abstract
Background and objective: Prevalence of atrial fibrillation is higher in hemodialysis patients as compared to the general population. Atrial electromechanical delay is known as a significant predictor of atrial fibrillation. In this study, we aimed to reveal the relationship between atrial electromechanical delay and attacks of atrial fibrillation. Materials and methods: The study included 77 hemodialysis patients over 18 years of age giving written consent to participate in the study. The patients were divided into two groups based on the results of 24-h Holter Electrocardiogram (Holter ECG) as the ones having attacks of atrial fibrillation and the others without any attack of atrial fibrillation. Standard echocardiographic measurements were taken from all patients. Additionally, atrial conduction times were measured by tissue Doppler technique and atrial electromechanical delays were calculated. Results: Intra- and interatrial electromechanical delay were found as significantly lengthened in the group of patients with attacks of atrial fibrillation (p = 0.03 and p < 0.001 respectively). The optimal cut-off time for interatrial electromechanical delay to predict atrial fibrillation was >21 ms with a specificity of 79.3% and a sensitivity of 73.7% (area under the curve 0.820; 95% confidence interval (CI), 0.716⁻0.898). In the multivariate logistic regression model, interatrial electromechanical delay (odds ratio = 1.230; 95% CI, 1.104⁻1.370; p < 0.001) and hypertension (odds ratio = 4.525; 95% CI, 1.042⁻19.651; p = 0.044) were also associated with atrial fibrillation after adjustment for variables found to be statistically significant in univariate analysis and correlated with interatrial electromechanical delay. Conclusions: Interatrial electromechanical delay is independently related with the attacks of atrial fibrillation detected on Holter ECG records in hemodialysis patients.
Collapse
Affiliation(s)
- Hakan Gunes
- Department of Cardiology, Sutcu Imam University, 46040 Kahramanmaras, Turkey.
| | - Abdullah Sokmen
- Department of Cardiology, Sutcu Imam University, 46040 Kahramanmaras, Turkey.
| | - Hakki Kaya
- Department of Cardiology, Cumhuriyet University, 58140 Sivas, Turkey.
| | - Ozkan Gungor
- Department of Nephrology, Sutcu Imam University, 46040 Kahramanmaras, Turkey.
| | - Murat Kerkutluoglu
- Department of Cardiology, Sutcu Imam University, 46040 Kahramanmaras, Turkey.
| | - Fatma Betul Guzel
- Department of Nephrology, Sutcu Imam University, 46040 Kahramanmaras, Turkey.
| | - Gulizar Sokmen
- Department of Cardiology, Sutcu Imam University, 46040 Kahramanmaras, Turkey.
| |
Collapse
|
313
|
The anti-inflammatory peptide Ac-SDKP: Synthesis, role in ACE inhibition, and its therapeutic potential in hypertension and cardiovascular diseases. Pharmacol Res 2018; 134:268-279. [DOI: 10.1016/j.phrs.2018.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/12/2018] [Accepted: 07/07/2018] [Indexed: 01/27/2023]
|
314
|
Kosmas CE, Silverio D, Tsomidou C, Salcedo MD, Montan PD, Guzman E. The Impact of Insulin Resistance and Chronic Kidney Disease on Inflammation and Cardiovascular Disease. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2018; 11:1179551418792257. [PMID: 30083062 PMCID: PMC6071166 DOI: 10.1177/1179551418792257] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/13/2022]
Abstract
There is extensive evidence showing that insulin resistance (IR) is associated
with chronic low-grade inflammation. Furthermore, IR has been shown to increase
the risk for cardiovascular disease (CVD), even in nondiabetic patients, and is
currently considered as a “nontraditional” risk factor contributing to CVD by
promoting hypertension, oxidative stress, endothelial dysfunction, dyslipidemia,
and type 2 diabetes mellitus. However, chronic kidney disease (CKD) is also
considered a state of low-grade inflammation. In addition, CKD is considered an
IR state and has been described as an independent risk factor for the
development of CVD, as even early-stage CKD is associated with an estimated 40%
to 100% increase in CVD risk. There is also strong evidence indicating that
inflammation per se plays a crucial role in both the initiation and progression
of CVD. Given the above, the combined effect of IR and CKD may significantly
increase the risk of inflammation and CVD. This review aims to focus on the
complex interplay between IR, CKD, inflammation, and CVD and will present and
discuss the current clinical and scientific data pertaining to the impact of IR
and CKD on inflammation and CVD.
Collapse
Affiliation(s)
- Constantine E Kosmas
- Division of Cardiology, Department of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Delia Silverio
- Cardiology Clinic, Cardiology Unlimited PC, New York, NY, USA
| | - Christiana Tsomidou
- Department of Medicine, General Clinic of Piraeus "Hippocrates", Piraeus, Greece
| | - Maria D Salcedo
- Cardiology Clinic, Cardiology Unlimited PC, New York, NY, USA
| | - Peter D Montan
- Cardiology Clinic, Cardiology Unlimited PC, New York, NY, USA
| | - Eliscer Guzman
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
315
|
Vahdat S. The complex effects of adipokines in the patients with kidney disease. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2018; 23:60. [PMID: 30181742 PMCID: PMC6091131 DOI: 10.4103/jrms.jrms_1115_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/12/2018] [Accepted: 04/29/2018] [Indexed: 12/27/2022]
Abstract
Kidney diseases are categorized as the highest prevalent ones with worldwide noticeable incidence. They cause accelerated cardiovascular diseases and noticeable mortalities. Adipose tissue and its messengers, adipokines, are reported to have the highest relationship with end-stage renal diseases or chronic kidney diseases. Over recent years, with shifting of scientists’ mindset from a simple overview of adipose tissue as a fat store to the complex paradigm of this issue as a multipotential secretory organ, the importance of studies on this tissue has emerged.
Collapse
Affiliation(s)
- Sahar Vahdat
- Isfahan Kidney Diseases Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
316
|
The Impact of Uremic Toxins on Cerebrovascular and Cognitive Disorders. Toxins (Basel) 2018; 10:toxins10070303. [PMID: 30037144 PMCID: PMC6071092 DOI: 10.3390/toxins10070303] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022] Open
Abstract
Individuals at all stages of chronic kidney disease (CKD) have a higher risk of developing cognitive disorders and dementia. Stroke is also highly prevalent in this population and is associated with a higher risk of neurological deterioration, in-hospital mortality, and poor functional outcomes. Evidence from in vitro studies and in vivo animal experiments suggests that accumulation of uremic toxins may contribute to the pathogenesis of stroke and amplify vascular damage, leading to cognitive disorders and dementia. This review summarizes current evidence on the mechanisms by which uremic toxins may favour the occurrence of cerebrovascular diseases and neurological complications in CKD.
Collapse
|
317
|
Zhang L, Wu JH, Otto JC, Gurley SB, Hauser ER, Shenoy SK, Nagi K, Brian L, Wertman V, Mattocks N, Lawson JH, Freedman NJ. Interleukin-9 mediates chronic kidney disease-dependent vein graft disease: a role for mast cells. Cardiovasc Res 2018; 113:1551-1559. [PMID: 29048463 DOI: 10.1093/cvr/cvx177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/29/2017] [Indexed: 12/18/2022] Open
Abstract
Aims Chronic kidney disease (CKD) is a powerful independent risk factor for cardiovascular events, including vein graft failure. Because CKD impairs the clearance of small proteins, we tested the hypothesis that CKD exacerbates vein graft disease by elevating serum levels of critical cytokines that promote vein graft neointimal hyperplasia. Methods and results We modelled CKD in C57BL/6 mice with 5/6ths nephrectomy, which reduced glomerular filtration rate by 60%, and we modelled vein grafting with inferior-vena-cava-to-carotid interposition grafting. CKD increased vein graft neointimal hyperplasia four-fold, decreased vein graft re-endothelialization two-fold, and increased serum levels of interleukin-9 (IL-9) five-fold. By quantitative immunofluorescence and histochemical staining, vein grafts from CKD mice demonstrated a ∼two-fold higher prevalence of mast cells, and a six-fold higher prevalence of activated mast cells. Concordantly, vein grafts from CKD mice showed higher levels of TNF and NFκB activation, as judged by phosphorylation of NFκB p65 on Ser536 and by expression of VCAM-1. Arteriovenous fistula veins from humans with CKD also showed up-regulation of mast cells and IL-9. Treating CKD mice with IL-9-neutralizing IgG reduced vein graft neointimal area four-fold, increased vein graft re-endothelialization ∼two-fold, and reduced vein graft total and activated mast cell levels two- and four-fold, respectively. Treating CKD mice with the mast cell stabilizer cromolyn reduced neointimal hyperplasia and increased re-endothelialization in vein grafts. In vitro, IL-9 promoted endothelial cell apoptosis but had no effect on smooth muscle cell proliferation. Conclusion CKD aggravates vein graft disease through mechanisms involving IL-9 and mast cell activation.
Collapse
Affiliation(s)
| | | | | | | | - Elizabeth R Hauser
- Biostatistics and Bioinformatics.,Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.,Cooperative Studies Program Epidemiology Center Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Sudha K Shenoy
- Cardiology, Department of Medicine.,Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Karim Nagi
- Cardiology, Department of Medicine.,Cell Biology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | - Neil J Freedman
- Cardiology, Department of Medicine.,Cell Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
318
|
Weiner DE, Park M, Tighiouart H, Joseph AA, Carpenter MA, Goyal N, House AA, Hsu CY, Ix JH, Jacques PF, Kew CE, Kim SJ, Kusek JW, Pesavento TE, Pfeffer MA, Smith SR, Weir MR, Levey AS, Bostom AG. Albuminuria and Allograft Failure, Cardiovascular Disease Events, and All-Cause Death in Stable Kidney Transplant Recipients: A Cohort Analysis of the FAVORIT Trial. Am J Kidney Dis 2018; 73:51-61. [PMID: 30037726 DOI: 10.1053/j.ajkd.2018.05.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/23/2018] [Indexed: 12/30/2022]
Abstract
RATIONALE & OBJECTIVE Cardiovascular disease (CVD) is common and overall graft survival is suboptimal among kidney transplant recipients. Although albuminuria is a known risk factor for adverse outcomes among persons with native chronic kidney disease, the relationship of albuminuria with cardiovascular and kidney outcomes in transplant recipients is uncertain. STUDY DESIGN Post hoc longitudinal cohort analysis of the Folic Acid for Vascular Outcomes Reduction in Transplantation (FAVORIT) Trial. SETTING & PARTICIPANTS Stable kidney transplant recipients with elevated homocysteine levels from 30 sites in the United States, Canada, and Brazil. PREDICTOR Urine albumin-creatinine ratio (ACR) at randomization. OUTCOMES Allograft failure, CVD, and all-cause death. ANALYTICAL APPROACH Multivariable Cox models adjusted for age; sex; race; randomized treatment allocation; country; systolic and diastolic blood pressure; history of CVD, diabetes, and hypertension; smoking; cholesterol; body mass index; estimated glomerular filtration rate (eGFR); donor type; transplant vintage; medications; and immunosuppression. RESULTS Among 3,511 participants with complete data, median ACR was 24 (Q1-Q3, 9-98) mg/g, mean eGFR was 49±18 (standard deviation) mL/min/1.73m2, mean age was 52±9 years, and median graft vintage was 4.1 (Q1-Q3, 1.7-7.4) years. There were 1,017 (29%) with ACR < 10mg/g, 912 (26%) with ACR of 10 to 29mg/g, 1,134 (32%) with ACR of 30 to 299mg/g, and 448 (13%) with ACR ≥ 300mg/g. During approximately 4 years, 282 allograft failure events, 497 CVD events, and 407 deaths occurred. Event rates were higher at both lower eGFRs and higher ACR. ACR of 30 to 299 and ≥300mg/g relative to ACR < 10mg/g were independently associated with graft failure (HRs of 3.40 [95% CI, 2.19-5.30] and 9.96 [95% CI, 6.35-15.62], respectively), CVD events (HRs of 1.25 [95% CI, 0.96-1.61] and 1.55 [95% CI, 1.13-2.11], respectively), and all-cause death (HRs of 1.65 [95% CI, 1.23-2.21] and 2.07 [95% CI, 1.46-2.94], respectively). LIMITATIONS No data for rejection; single ACR assessment. CONCLUSIONS In a large population of stable kidney transplant recipients, elevated baseline ACR is independently associated with allograft failure, CVD, and death. Future studies are needed to evaluate whether reducing albuminuria improves these outcomes.
Collapse
Affiliation(s)
| | - Meyeon Park
- Division of Nephrology, University of California, San Francisco, San Francisco, CA
| | | | - Alin A Joseph
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Myra A Carpenter
- Collaborative Studies Coordinating Center, University of North Carolina, Chapel Hill, NC
| | - Nitender Goyal
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Andrew A House
- Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California, San Francisco, San Francisco, CA
| | - Joachim H Ix
- Division of Nephrology-Hypertension, University of California, San Diego, San Diego, CA
| | - Paul F Jacques
- Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Clifton E Kew
- Division of Nephrology, University of Alabama, Birmingham, AL
| | - S Joseph Kim
- Division of Nephrology and the Kidney Transplant Program, Toronto General Hospital, Toronto, Ontario, Canada
| | - John W Kusek
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | | | - Marc A Pfeffer
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | | | - Matthew R Weir
- Division of Nephrology, University of Maryland, Baltimore, MD
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Andrew G Bostom
- Division of Hypertension and Kidney Diseases, Rhode Island Hospital, Providence, RI
| |
Collapse
|
319
|
Intestinal Barrier Function in Chronic Kidney Disease. Toxins (Basel) 2018; 10:toxins10070298. [PMID: 30029474 PMCID: PMC6071212 DOI: 10.3390/toxins10070298] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/16/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022] Open
Abstract
The kidneys are key contributors to body homeostasis, by virtue of controlled excretion of excessive fluid, electrolytes, and toxic waste products. The syndrome of uremia equals the altered physiology due to irreversible loss of kidney function that is left uncorrected for, despite therapeutic intervention(s). The intestines and its microbial content are prime contributors to this syndrome. The intestinal barrier separates the self (or the so-called “milieu intérior”) from the environment. In the large intestine, the intestinal barrier keeps apart human physiology and the microbiota. The enterocytes and the extracellular mucin layer functions form a complex multilayered structure, facilitating complex bidirectional metabolic and immunological crosstalk. The current review focuses on the intestinal barrier in chronic kidney disease (CKD). Loss of kidney function results in structural and functional alterations of the intestinal barrier, contribution to the syndrome of uremia.
Collapse
|
320
|
Abstract
Chronic, low-grade inflammation is a common comorbid condition in chronic kidney disease (CKD), and particularly in chronic dialysis patients. In this review, we consider the question of whether inflammation affects outcomes in dialysis patients. Levels of proinflammatory cytokines, as well as C-reactive protein, are elevated in chronic dialysis patients. Multiple factors likely contribute to chronic inflammatory activation in kidney disease patients including the uremic milieu, lifestyle and epigenetic influences, infectious and thrombotic events, the dialysis process, and dysbiosis. Increased inflammatory markers in both CKD and chronic dialysis patients are associated with adverse clinical outcomes including all-cause mortality, cardiovascular events, kidney disease progression, protein energy wasting and diminished motor function, cognitive impairment, as well as other adverse consequences including CKD-mineral and bone disorder, anemia, and insulin resistance. Strategies that have been shown to reduce chronic systemic inflammation in CKD and chronic dialysis patients include both pharmacological and nonpharmacological interventions. However, despite evidence that systemic inflammatory markers can be lowered in kidney disease patients treated with various strategies, evidence that this improves clinical outcomes is largely unavailable and represents an important future research direction. Overall, there is strong observational evidence that inflammation is high in chronic dialysis patients and that this is independently associated with numerous adverse clinical outcomes. Targeting inflammation represents a potentially novel and attractive strategy if it can indeed improve adverse outcomes common in this population.
Collapse
|
321
|
Rasmussen DGK, Hansen TW, von Scholten BJ, Nielsen SH, Reinhard H, Parving HH, Tepel M, Karsdal MA, Jacobsen PK, Genovese F, Rossing P. Higher Collagen VI Formation Is Associated With All-Cause Mortality in Patients With Type 2 Diabetes and Microalbuminuria. Diabetes Care 2018; 41:1493-1500. [PMID: 29643059 DOI: 10.2337/dc17-2392] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/26/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 2 diabetes is a common risk factor for the development of chronic kidney disease (CKD). Enhanced de novo collagen type VI (COL VI) formation has been associated with renal fibrosis and CKD. We investigated the hypothesis that PRO-C6, a product specifically generated during COL VI formation, is prognostic for adverse outcomes in patients with type 2 diabetes and microalbuminuria. RESEARCH DESIGN AND METHODS In a prospective, observational study, we measured PRO-C6 in the serum (S-PRO-C6) and urine (U-PRO-C6) of 198 patients with type 2 diabetes and microalbuminuria without symptoms of coronary artery disease. Patients were followed for a median of 6.5 years, and end points were a composite of cardiovascular events (n = 38), all-cause mortality (n = 26), and reduction of estimated glomerular filtration rate (eGFR) of >30% (disease progression [n = 42]). Cox models were unadjusted and adjusted for the conventional risk factors of sex, age, BMI, systolic blood pressure, LDL cholesterol, smoking, HbA1c, plasma creatinine, and urinary albumin excretion rate. RESULTS Doubling of S-PRO-C6 increased hazards for cardiovascular events (hazard ratio 3.06 [95% CI 1.31-7.14]), all-cause mortality (6.91 [2.96-16.11]), and disease progression (4.81 [1.92-12.01]). Addition of S-PRO-C6 to a model containing conventional risk factors improved relative integrated discrimination by 22.5% for cardiovascular events (P = 0.02), 76.8% for all-cause mortality (P = 0.002), and 53.3% for disease progression (P = 0.004). U-PRO-C6 was not significantly associated with any of the outcomes. CONCLUSIONS S-PRO-C6 generated during COL VI formation predicts cardiovascular events, all-cause mortality, and disease progression in patients with type 2 diabetes and microalbuminuria.
Collapse
Affiliation(s)
- Daniel G K Rasmussen
- Nordic Bioscience, Herlev, Denmark .,Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | | | | | - Signe H Nielsen
- Nordic Bioscience, Herlev, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | | | - Martin Tepel
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.,Department of Nephrology, Odense University Hospital, Odense, Denmark
| | | | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
322
|
Yu TY, Kim HY, Lee JM, Lee DH, Cho CG. Association between Bone Mineral Density and Albuminuria: Cross-Sectional Analysis of Data from the 2011 Korea National Health and Nutrition Examination Survey V-2. Endocrinol Metab (Seoul) 2018; 33:211-218. [PMID: 29766678 PMCID: PMC6021310 DOI: 10.3803/enm.2018.33.2.211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/25/2018] [Accepted: 02/22/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Albuminuria is known to be independently associated with progression of renal and cardiovascular disease. However, little is known regarding the exact relationship between albuminuria and bone mineral density (BMD). The aim of this population-based study conducted in Korea was to identify the association between albuminuria and BMD. METHODS We performed a cross-sectional analysis of data from the Korea National Health and Nutrition Examination Survey (KNHANES V-2) 2011. BMD was measured for total hip (TH), femur neck (FN), and lumbar spine (LS). Analysis of covariance was used to compare BMD levels between the groups at the TH, FN, and LS sites, after adjusting for age. Separate analyses were performed according to sex; women were divided into two groups according to menopausal status and each group was subdivided into three according to urine albumin-to-creatinine ratio (level 1, <30 mg/g; level 2, 30 to 299 mg/g; level 3, ≥300 mg/g). RESULTS Data on a total of 1,831 adults (857 men and 974 women) were analyzed. In postmenopausal women, after adjusting for age, BMD of TH tended to decrease as levels of albuminuria increased (0.767±0.117, 0.757±0.129, 0.752±0.118, respectively; P=0.040). However, there was no significant difference in BMD according to albuminuria level in premenopausal women and men. CONCLUSION Level of albuminuria was closely related with BMD of TH in postmenopausal women, after adjusting for age, but there was no significant relationship between albuminuria and BMD in premenopausal women and men.
Collapse
Affiliation(s)
- Tae Yang Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Korea
| | - Ha Young Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wonkwang University Sanbon Hospital, Wonkwang University School of Medicine, Gunpo, Korea
| | - Jeong Mi Lee
- Department of Public Health, Wonkwang University School of Medicine, Iksan, Korea
| | - Dae Ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.
| | - Chung Gu Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Korea.
| |
Collapse
|
323
|
Rossaint J, Unruh M, Zarbock A. Fibroblast growth factor 23 actions in inflammation: a key factor in CKD outcomes. Nephrol Dial Transplant 2018; 32:1448-1453. [PMID: 27659127 DOI: 10.1093/ndt/gfw331] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
During chronic kidney disease (CKD), bone mineral metabolism is disturbed owing in part to the endogenous hormone fibroblast growth factor 23 (FGF23). Elevated FGF23 levels are seen in CKD patients. Current research has demonstrated that FGF23 directly modulates the immune response and host defense to bacterial infections. FGF23 also impairs the activation and recruitment of neutrophils, which are the main immune effector cells required for host defense against bacterial infections. In addition, while FGF23 levels reduce leukocyte recruitment and functions, inflammatory conditions may also-in a reverse fashion-contribute to elevated FGF23 levels in the circulation. In this context, altered hypoxia inducible factor 1α signaling and iron metabolism may contribute to intact FGF23 (iFGF23) production. This review examines evidence on the role of FGF23 in inflammation, immune cell function and recruitment as well as the regulation of FGF23 during inflammation and the clinical implications of this process for the immune system in individuals with CKD. Clinical observations and laboratory investigations indicate an important role of FGF23 in directly modulating leukocyte activation and recruitment behavior with consequences on host defense against bacterial infections. This novel observation may in part explain the increased infectious risk among patients with CKD. However, studies of FGF23 neutralization also revealed increased mortality after sustained administration over several weeks in rats. Thus, therapeutic interventions targeting FGF23 must be carefully evaluated.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Mark Unruh
- Division of Nephrology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA.,Section of Neprology, New Mexico Veteran Affairs Hospital, Albuquerque, NM, USA
| | - Alexander Zarbock
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
324
|
Mlodawska E, Lopatowska P, Malyszko J, Banach M, Sobkowicz B, Covic A, Tomaszuk-Kazberuk A. Atrial fibrillation in dialysis patients: is there a place for non-vitamin K antagonist oral anticoagulants? Int Urol Nephrol 2018; 50:1633-1642. [PMID: 29785661 DOI: 10.1007/s11255-018-1877-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/18/2018] [Indexed: 01/03/2023]
Abstract
Atrial fibrillation (AF) occurs approximately in 3% of general population, with greater prevalence in elderly. Non-vitamin K-dependent oral anticoagulant agents (NOACs) according to the current European guidelines are recommended for patients with AF at high risk for stroke as a first-choice treatment. NOACs are not inferior to warfarin or some of them are better than warfarin in reducing the rate of ischemic stroke. Moreover, they significantly reduce the rate of intracranial hemorrhages, major bleedings, and mortality compared with warfarin. Nevertheless according to ESC guidelines, NOACs are not recommended in patients with creatinine clearance < 30 mL/min. Observational studies provide contradictive data. Only few new trials are ongoing. Therefore, it is not clear if NOACs should be in the future prescribed to patients with advanced CKD and those on dialysis. Moreover, the risk of stroke and bleeding is much higher in such population than in patients without end-stage renal disease (ESRD). The authors provide data on pros and cons of use of NOACs in ESRD patients with AF.
Collapse
Affiliation(s)
- Elzbieta Mlodawska
- Department of Cardiology, Medical University in Bialystok, Białystok, Poland
| | - Paulina Lopatowska
- Department of Cardiology, Medical University in Bialystok, Białystok, Poland
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Medicine, Warsaw Medical University, Warsaw, Poland
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| | - Bożena Sobkowicz
- Department of Cardiology, Medical University in Bialystok, Białystok, Poland
| | - Adrian Covic
- Nephrology Clinic, Dialysis and Renal Transplant Center - 'C.I. Parhon' University Hospital, and 'Grigore T. Popa' University of Medicine, Iasi, Romania
| | - Anna Tomaszuk-Kazberuk
- Department of Cardiology, Medical University in Bialystok, Białystok, Poland. .,Department of Cardiology, University Hospital in Bialystok, ul. Skłodowskiej-Curie 24A, 15-276, Białystok, Poland.
| |
Collapse
|
325
|
Wolley M, Jardine M, Hutchison CA. Exploring the Clinical Relevance of Providing Increased Removal of Large Middle Molecules. Clin J Am Soc Nephrol 2018; 13:805-814. [PMID: 29507008 PMCID: PMC5969479 DOI: 10.2215/cjn.10110917] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Dialysis technologies have continued to advance over recent decades; however, these advancements have not always been met with improved patient outcomes. In part, the high morbidity and mortality associated with dialysis have been attributed to a group of uremic toxins, which are described as "difficult to remove." With a new generation of hemodialysis membranes now making meaningful clearance of these molecules possible, it is an apt time to review the clinical relevance of these middle molecules. Our review describes the developments in membrane technology that enable the removal of large middle molecules (molecular mass >15 kD) that is limited with high-flux dialysis membranes. Of the known 58 middle molecules, a literature search identified 27 that have molecular mass >15 kD. This group contains cytokines, adipokines, hormones, and other proteins. These molecules are implicated in chronic inflammation, atherosclerosis, structural heart disease, and secondary immunodeficiency in the literature. Single-center safety and efficacy studies have identified that use of these membranes in maintenance dialysis populations is associated with limited loss of albumin and increased clearance of large middle molecules. Larger, robustly conducted, multicenter studies are now evaluating these findings. After completion of these safety and efficacy studies, the perceived clinical benefits of providing clearance of large middle molecules must be assessed in rigorously conducted, randomized clinical studies.
Collapse
Affiliation(s)
- Martin Wolley
- Department of Renal Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Meg Jardine
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Renal Medicine, Concord Repatriation General Hospital and University of Sydney, Sydney, New South Wales, Australia; and
| | - Colin A. Hutchison
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Department of Medicine, Hawke’s Bay District Health Board, Hastings, New Zealand
| |
Collapse
|
326
|
Phan T, Mcmillan R, Skiadopoulos L, Walborn A, Hoppensteadt D, Fareed J, Bansal V. Elevated extracellular nucleosomes and their relevance to inflammation in stage 5 chronic kidney disease. INT ANGIOL 2018; 37:419-426. [PMID: 29644836 DOI: 10.23736/s0392-9590.18.03987-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Chronic kidney disease is a disorder characterized by a pro-inflammatory state that is associated with increased morbidity and mortality. Endogenous damage-associated molecular patterns, including nucleosomes, may contribute to this persistent inflammation. The aim of this study was to profile and evaluate the clinical significance of circulating nucleosomes in patients with Stage 5 chronic kidney disease (CKD5) on hemodialysis (HD). METHODS Under institutional review board approval, plasma samples were collected from 90 CKD5-HD patients (45 male and 45 female) prior to hemodialysis. Normal human plasma samples (25 male and 25 female) were used as a control group. Commercial enzyme-linked immunosorbent and colorimetric assays were used to profile nucleosome levels and biochemical markers of kidney injury, inflammation, thrombosis, and renal function in CKD5-HD and control groups. Clinical laboratory parameters were documented from the electronical medical record and correlated to nucleosome levels in the CKD5-HD cohort. RESULTS In comparison to healthy volunteers, the plasma from CKD5-HD patients exhibited markedly elevated nucleosomes (P<0.0001). Furthermore, nucleosome levels correlated with WBC count (P=0.025, R=0.243) and CRP (P=0.019, R=0.266) levels. No correlation was found between nucleosomes and the other parameters studied. CONCLUSIONS Our findings indicate extracellular nucleosomes are significantly elevated in CKD5-HD, suggesting increased cell death and/or inflammation. The observed correlations between nucleosomes and parameters of inflammation is suggestive of a complex, systemic inflammatory process underlying renal deterioration, consistent with the literature. Thus, nucleosomes may play a role in the pathogenesis and outcome of CKD5-HD.
Collapse
Affiliation(s)
- Trung Phan
- Stritch School of Medicine, Loyola University of Chicago, Maywood, IL, USA
| | - Ryan Mcmillan
- Stritch School of Medicine, Loyola University of Chicago, Maywood, IL, USA
| | | | - Amanda Walborn
- Stritch School of Medicine, Loyola University of Chicago, Maywood, IL, USA
| | - Debra Hoppensteadt
- Unit of Hemostasis and Thrombosis, Department of Pathology, Loyola University Medical Center, Maywood, IL, USA
| | - Jawed Fareed
- Unit of Hemostasis and Thrombosis, Department of Pathology, Loyola University Medical Center, Maywood, IL, USA -
| | - Vinod Bansal
- Unit of Nephrology, Department of Internal Medicine, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
327
|
Translational science in albuminuria: a new view of de novo albuminuria under chronic RAS suppression. Clin Sci (Lond) 2018; 132:739-758. [DOI: 10.1042/cs20180097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/09/2018] [Accepted: 03/22/2018] [Indexed: 12/29/2022]
Abstract
The development of de novo albuminuria during chronic renin–angiotensin system (RAS) suppression is a clinical entity that remains poorly recognized in the biomedical literature. It represents a clear increment in global cardiovascular (CV) and renal risk that cannot be counteracted by RAS suppression. Although not specifically considered, it is clear that this entity is present in most published and ongoing trials dealing with the different forms of CV and renal disease. In this review, we focus on the mechanisms promoting albuminuria, and the predictors and new markers of de novo albuminuria, as well as the potential treatment options to counteract the excretion of albumin. The increase in risk that accompanies de novo albuminuria supports the search for early markers and predictors that will allow practising physicians to assess and prevent the development of de novo albuminuria in their patients.
Collapse
|
328
|
Use of oral anticoagulants in patients with atrial fibrillation and renal dysfunction. Nat Rev Nephrol 2018; 14:337-351. [PMID: 29578207 DOI: 10.1038/nrneph.2018.19] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atrial fibrillation (AF) and chronic kidney disease (CKD) are increasingly prevalent in the general population and share common risk factors such as older age, hypertension and diabetes mellitus. The presence of CKD increases the risk of incident AF, and, likewise, AF increases the risk of CKD development and/or progression. Both conditions are associated with substantial thromboembolic risk, but patients with advanced CKD also exhibit a paradoxical increase in bleeding risk. In the landmark randomized clinical trials that compared non-vitamin K antagonist oral anticoagulants (NOACs) with warfarin for thromboprophylaxis in patients with AF, the efficacy and safety of NOACs in patients with mild-to-moderate CKD were similar to those in patients without CKD. Dose adjustment of NOACs as per the prescribing label is required in this population. Owing to limited trial data, evidence-based recommendations for the management of patients with AF and severe CKD or end-stage renal disease on dialysis are lacking. Observational cohort studies have reported conflicting results, and the management of these particularly vulnerable patients remains challenging and requires careful assessment of stroke and bleeding risk and, where appropriate, use of warfarin with good-quality anticoagulation control.
Collapse
|
329
|
Assessment of physical performance and body composition in male renal transplant patients. J Nephrol 2018; 31:613-620. [PMID: 29551009 DOI: 10.1007/s40620-018-0483-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/22/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Renal transplant (RTX) recipients seem to experience a better quality of life compared to dialysis patients. However, the factors responsible for this positive effect are not completely defined. Conceivably, a change in the physical performance of these patients could play a role. METHODS To assess this, we measured: (1) waist circumference, fat mass and appendicular fat-free mass (aFFM) by dual-energy X-ray densitometry, (2) physical performance with the Short Physical Performance Battery, and (3) muscle strength with the handgrip test, in 59 male RTX, 11 chronic kidney disease in conservative treatment (CKD) and 10 peritoneal dialysis (PD) patients. RESULTS Surprisingly, anthropometric characteristics and body composition were similar among the three groups. However, despite a low aFFM, muscle strength was higher in stable RTX recipients > 5 years after transplantation than in dialyzed patients. Instead, CKD (wait-listed for RTX) had similar muscle strength to RTX patients. Waist circumference in RTX recipients showed a redistribution of body fat with increased central adipose tissue allocation compared to PD. At linear regression analysis, age, weight, height, aFFM, hemoglobin and transplant age were independent predictors of handgrip strength, explaining about 37% of the variance. Age and transplant age accounted for 18 and 12% of variance, respectively. CONCLUSIONS Our study demonstrates, for the first time, that clinically stable RTX recipients have greater muscle strength than dialyzed patients and suggests that the handgrip test could be an effective and easy-to-perform tool to assess changes in physical performance in this large patient population.
Collapse
|
330
|
Kim HJ, Min JY, Seo YS, Min KB. Association between exposure to ambient air pollution and renal function in Korean adults. Ann Occup Environ Med 2018; 30:14. [PMID: 29507730 PMCID: PMC5831208 DOI: 10.1186/s40557-018-0226-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Ambient air pollution has a negative effect on many diseases, such as cardiovascular and respiratory diseases. Recent studies have reported a relationship between air pollution and renal function, but the results were limited to exposure to particulate matter (PM). This study was to identify associations between various air pollutants and renal function among Korean adults. Methods Nationwide survey data for a total of 24,407 adults were analyzed. We calculated the estimated glomerular filtration rate (eGFR) for each individual to assess their renal function and used this to categorize those with chronic kidney disease (CKD). To evaluate exposure to ambient air pollution, we used the annual mean concentrations of four ambient air pollutants: PM with an aerodynamic diameter ≤ 10 μm (PM10), nitrogen dioxide (NO2), sulfur dioxide (SO2), and carbon monoxide (CO). Results We identified significant inverse relationships between the air pollutants PM10 and NO2 and eGFR in all statistical adjustment models (all p < 0.05). In the full covariate model, interquartile range increases in the annual mean concentrations of PM10 and NO2 were associated with decreases in eGFR levels of 0.46 (95% CI = − 0.87, − 0.04) and 0.85 (95% CI = − 1.40, − 0.30), respectively. Three of the ambient air pollutants were significantly related to an increased risk of CKD in the unadjusted model (p < 0.0001), but all significant associations disappeared after adjusting for covariates (all p > 0.05). Conclusions Exposures to PM10 and NO2 were significantly associated with decreases in eGFR levels, but not CKD, in Korean adults.
Collapse
Affiliation(s)
- Hyun-Jin Kim
- 1Institute of Health and Environment, Seoul National University, Seoul, 08826 Republic of Korea
| | - Jin-Young Min
- 2Institute of Health and Environment, Seoul National University, Seoul, 08826 Republic of Korea
| | - Yong-Seok Seo
- 3Institute of Environmental Research, Kangwon National University, Chuncheon, 24341 Gangwon-do Republic of Korea
| | - Kyoung-Bok Min
- 4Department of Preventive Medicine, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| |
Collapse
|
331
|
Wei J, Zhang J, Wang L, Cha BJ, Jiang S, Liu R. A new low-nephron CKD model with hypertension, progressive decline of renal function, and enhanced inflammation in C57BL/6 mice. Am J Physiol Renal Physiol 2018; 314:F1008-F1019. [PMID: 29412703 DOI: 10.1152/ajprenal.00574.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is a major health issue in the US. The typical five-sixths nephrectomy (typical 5/6 NX) is a widely used experimental CKD model. However, the typical 5/6 NX model is hypertensive in rats but strain dependent in mice. In particular, C57BL/6 mice with the typical 5/6 NX exhibits normal blood pressure and well-preserved renal function. The goal of the present study was to create a new hypertensive CKD model in C57BL/6 mice. We first characterized the vascular architecture originated from each renal artery branch by confocal laser-scanning microscopy with fluorescent lectin. Then, a novel 5/6 NX-BL model was generated by uninephrectomy combined with 2/3 renal infarction via a ligation of upper renal artery branch on the contralateral kidney. Compared with 5/6 NX-C, the 5/6 NX-BL model exhibited elevated mean arterial pressure (137.6 ± 13.9 vs. 104.7 ± 8.2 mmHg), decreased glomerular filtration rate (82.9 ± 19.2 vs. 125.0 ± 13.9 µl/min) with a reciprocal increase in plasma creatinine (0.31 ± 0.03 vs. 0.19 ± 0.04 mg/dl), and significant renal injury as assessed by proteinuria, histology with light, and transmission electron microscopy. In addition, inflammatory status, as indicated by the level of proinflammatory cytokine TNFα and the leukocyte counts, was significantly upregulated in 5/6 NX-BL compared with the 5/6 NX-C. In summary, we developed a new hypertensive CKD model in C57BL/6 mice with 5/6 renal mass reduction by uninephrectomy and upper renal artery branch ligation on the contralateral kidney. This 5/6 NX-BL model exhibits an infarction zone-dependent hypertension and progressive deterioration of the renal function accompanied by enhanced inflammatory response.
Collapse
Affiliation(s)
- Jin Wei
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Jie Zhang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Byeong Jake Cha
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Shan Jiang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| |
Collapse
|
332
|
Haddad Kashani H, Seyed Hosseini E, Nikzad H, Soleimani A, Soleimani M, Tamadon MR, Keneshlou F, Asemi Z. The Effects of Vitamin D Supplementation on Signaling Pathway of Inflammation and Oxidative Stress in Diabetic Hemodialysis: A Randomized, Double-Blind, Placebo-Controlled Trial. Front Pharmacol 2018; 9:50. [PMID: 29456507 PMCID: PMC5801479 DOI: 10.3389/fphar.2018.00050] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/15/2018] [Indexed: 01/02/2023] Open
Abstract
Objective: This study was carried out to determine the effects of vitamin D supplementation on signaling pathway of inflammation and oxidative stress in diabetic hemodialysis (HD) patients. Methods: This randomized double-blind placebo-controlled clinical trial was conducted among 60 diabetic HD patients. Subjects were randomly allocated into two groups to intake either vitamin D supplements at a dosage of 50,000 IU (n = 30) or placebo (n = 30) every 2 weeks for 12 weeks. Gene expression of inflammatory cytokines and biomarkers of oxidative stress were assessed in peripheral blood mononuclear cells (PBMCs) of diabetic HD patients with RT-PCR method. Results: Results of RT-PCR indicated that after the 12-week intervention, compared to the placebo, vitamin D supplementation downregulated gene expression of interleukin (IL)-1β (P = 0.02), tumor necrosis factor alpha (TNF-α) (P = 0.02) and interferon gamma (IFN-γ) (P = 0.03) in PBMCs of diabetic HD patients. Additionally, vitamin D supplementation, compared to the placebo, downregulated gene expression of transforming growth factor beta (TGF-β) (P = 0.04), protein kinase C (PKC) (P = 0.001), and mitogen-activated protein kinases 1 (MAPK1) (P = 0.02) in PBMCs of diabetic HD patients. Although not significant, vitamin D supplementation let to a reduction of nuclear factor kappa B (NF-kB) (p = 0.75) expression in PBMCs isolated from diabetic patients compared to the placebo group. There was no statistically significant change following supplementation with vitamin D on gene expression of interleukin (IL)-4, IL-6, and vascular endothelial growth factor (VEGF) in PBMCs of diabetic HD patients. Conclusions: Overall, we found that vitamin D supplementation for 12 weeks among diabetic HD patients had beneficial effects on few gene expression related to inflammation and oxidative stress. Clinical trial registration: IRCT201701035623N101. Registered on January 8, 2017.
Collapse
Affiliation(s)
- Hamed Haddad Kashani
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Elahe Seyed Hosseini
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hossein Nikzad
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Soleimani
- Department of Internal Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Soleimani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Reza Tamadon
- Department of Internal Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fariba Keneshlou
- Department of Urology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
333
|
Massicotte-Azarniouch D, Kuwornu JP, Carrero JJ, Lam NN, Molnar AO, Zimmerman D, McCallum MK, Garg AX, Sood MM. Incident Atrial Fibrillation and the Risk of Congestive Heart Failure, Myocardial Infarction, End-Stage Kidney Disease, and Mortality Among Patients With a Decreased Estimated GFR. Am J Kidney Dis 2018; 71:191-199. [DOI: 10.1053/j.ajkd.2017.08.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/09/2017] [Indexed: 11/11/2022]
|
334
|
Sharp CN, Doll MA, Megyesi J, Oropilla GB, Beverly LJ, Siskind LJ. Subclinical kidney injury induced by repeated cisplatin administration results in progressive chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F161-F172. [PMID: 29384415 DOI: 10.1152/ajprenal.00636.2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cisplatin is used to treat many solid cancers, but its dose-limiting side effect is nephrotoxicity, causing acute kidney injury in 30% of patients. Previously, we have developed a mouse model that better recapitulates the cisplatin dosing regimen humans receive and found that repeated dosing of cisplatin induces interstitial renal fibrosis. Chronic kidney disease is progressive and is characterized by chronic inflammation, worsening interstitial fibrosis, development of glomerulosclerosis, and endothelial dysfunction. To determine if damage caused by repeated cisplatin dosing results in bona fide chronic kidney disease, mice were treated with our repeated dosing regimen and then aged for 6 mo. These mice had progressive, chronic inflammation and worsened interstitial fibrosis compared with mice euthanized after day 24. Mice aged for 6 mo developed glomerular pathologies, and endothelial dysfunction was persistent. Mice treated with only two doses of cisplatin had little inflammation or kidney damage. Thus repeated dosing of cisplatin causes long-term effects that are characteristic of chronic kidney disease. This translational mouse model of cisplatin injury may better represent the 70% of patients that do not develop clinical acute kidney injury and can be used to identify both biomarkers for early injury, as well as novel therapeutic targets for the prevention of cisplatin-induced chronic kidney disease.
Collapse
Affiliation(s)
- Cierra N Sharp
- Department of Pharmacology and Toxicology, University of Louisville , Louisville, Kentucky
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville , Louisville, Kentucky
| | - Judit Megyesi
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System , Little Rock, Arkansas
| | | | - Levi J Beverly
- James Graham Brown Cancer Center, University of Louisville , Louisville, Kentucky.,Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville , Louisville, Kentucky.,James Graham Brown Cancer Center, University of Louisville , Louisville, Kentucky
| |
Collapse
|
335
|
Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018. [PMID: 29467962 DOI: 10.1832/oncotarget.23208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds. These factors may induce acute and/or chronic inflammatory responses in the heart, pancreas, liver, kidney, lung, brain, intestinal tract and reproductive system, potentially leading to tissue damage or disease. Both infectious and non-infectious agents and cell damage activate inflammatory cells and trigger inflammatory signaling pathways, most commonly the NF-κB, MAPK, and JAK-STAT pathways. Here, we review inflammatory responses within organs, focusing on the etiology of inflammation, inflammatory response mechanisms, resolution of inflammation, and organ-specific inflammatory responses.
Collapse
Affiliation(s)
- Linlin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| |
Collapse
|
336
|
Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018; 9:7204-7218. [PMID: 29467962 PMCID: PMC5805548 DOI: 10.18632/oncotarget.23208] [Citation(s) in RCA: 2701] [Impact Index Per Article: 385.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds. These factors may induce acute and/or chronic inflammatory responses in the heart, pancreas, liver, kidney, lung, brain, intestinal tract and reproductive system, potentially leading to tissue damage or disease. Both infectious and non-infectious agents and cell damage activate inflammatory cells and trigger inflammatory signaling pathways, most commonly the NF-κB, MAPK, and JAK-STAT pathways. Here, we review inflammatory responses within organs, focusing on the etiology of inflammation, inflammatory response mechanisms, resolution of inflammation, and organ-specific inflammatory responses.
Collapse
Affiliation(s)
- Linlin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| |
Collapse
|
337
|
Ishigami J, Grams ME, Naik RP, Caughey MC, Loehr LR, Uchida S, Coresh J, Matsushita K. Hemoglobin, Albuminuria, and Kidney Function in Cardiovascular Risk: The ARIC (Atherosclerosis Risk in Communities) Study. J Am Heart Assoc 2018; 7:JAHA.117.007209. [PMID: 29330257 PMCID: PMC5850152 DOI: 10.1161/jaha.117.007209] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Reduced estimated glomerular filtration rate (eGFR) and elevated urinary albumin-to-creatinine ratio (ACR) individually increase risk of cardiovascular disease (CVD). We hypothesized that these associations are stronger among people with abnormal (both low and high) hemoglobin levels. METHODS AND RESULTS Using 5801 participants with available hemoglobin measures of the ARIC (Atherosclerosis Risk in Community) study in 1996-1998, we explored the cross-sectional association of eGFR and ACR with hemoglobin levels and their longitudinal associations with CVD (heart failure, coronary heart disease, and stroke) risk through 2013. At baseline, 8.8% had anemia (<13 g/dL in men and <12 g/dL in women) and 7.2% had high hemoglobin (≥16 g/dL in men and ≥15 g/dL in women). The adjusted prevalence ratio of anemia was 2.12 (95% confidence interval, 1.59-2.82) for eGFR 30 to 59 compared with ≥90 mL/min per 1.73 m2 and 1.45 (1.07-1.95) for ACR ≥30 compared with <10 mg/g. ACR ≥30 mg/g was also associated with high hemoglobin (prevalence ratio, 1.57 [1.12-2.19] compared with <10 mg/g). During follow-up, there were 1069 incident CVDs among 5098 CVD-free participants at baseline. In multivariable Cox models, lower eGFR, higher ACR, and anemia were each independently associated with CVD risk, with the association of low eGFR being slightly stronger in anemia (P-for-interaction, 0.072). There was no hemoglobin-ACR interaction; however, when CVD subtypes were analyzed separately, risk of coronary heart disease and stroke associated with high ACR was slightly stronger in high hemoglobin (P-for-interaction, 0.074). CONCLUSIONS Kidney function, albuminuria, and anemia were correlated and independently associated with CVD risk. Correlation and potential interaction for atherosclerotic CVD between albuminuria and high hemoglobin deserve further investigation.
Collapse
Affiliation(s)
- Junichi Ishigami
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Morgan E Grams
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Rakhi P Naik
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Melissa C Caughey
- Department of Medicine, University of North Carolina at Chapel Hill, NC
| | - Laura R Loehr
- Department of Epidemiology, University of North Carolina at Chapel Hill, NC
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
338
|
Hepatocyte nuclear factors as possible C-reactive protein transcriptional inducer in the liver and white adipose tissue of rats with experimental chronic renal failure. Mol Cell Biochem 2018; 446:11-23. [PMID: 29330688 PMCID: PMC6096500 DOI: 10.1007/s11010-018-3268-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/04/2018] [Indexed: 12/11/2022]
Abstract
Inflammation related to chronic kidney disease (CKD) is an important clinical problem. We recently determined that hepatocyte nuclear factor 1α (HNF1α) was upregulated in the livers of chronic renal failure (CRF) rats—experimental model of CKD. Considering that the promoter region of gene encoding C-reactive protein (CRP) contains binding sites for HNF1α and that the loss-of-function mutation in the Hnfs1α leads to significant reduction in circulating CRP levels, we hypothesized that HNF1α can activate the Crp in CRF rats. Here, we found coordinated upregulation of genes encoding CRP, interleukin-6 (IL-6), HNF1α, and HNF4α in the livers and white adipose tissue (WAT) of CRF rats, as compared to the pair-fed and control animals. This was accompanied by elevated serum levels of CRP and IL-6. CRP and HNFs’ mRNA levels correlated positively with CRP and HNFs’ protein levels in the liver and WAT. Similar upregulation of the Crp, Il-6, and Hnfs in the liver and WAT and increased serum CRP and IL-6 concentrations were found in lipopolysaccharide (LPS)-induced systemic inflammation in rats. Moreover, silencing HNF1α in HepG2 cells by small interfering RNA led to decrease in CRP mRNA levels. Our results suggests that (a) HNFs act in concert with IL-6 in the upregulation of CRP production by the liver and WAT, leading to an increase in circulating CRP concentration in CRF rats and (b) CRF-related inflammation plays an important role in the upregulation of genes that encode HNFs and CRP in the liver and WAT of CRF rats.
Collapse
|
339
|
Esmeijer K, Geleijnse JM, Giltay EJ, Stijnen T, Dekker FW, de Fijter JW, Kromhout D, Hoogeveen EK. Body-fat indicators and kidney function decline in older post-myocardial infarction patients: The Alpha Omega Cohort Study. Eur J Prev Cardiol 2018; 25:90-99. [PMID: 29095051 PMCID: PMC5724586 DOI: 10.1177/2047487317739986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/10/2017] [Indexed: 11/24/2022]
Abstract
Background Obesity increases risk of hypertension and diabetes, the leading causes of end-stage renal disease. The effect of obesity on kidney function decline in stable post-myocardial infarction patients is poorly documented. This relation was investigated in a large cohort of older post-myocardial infarction patients. Design Data were analysed from 2410 post-myocardial infarction patients in the Alpha Omega Trial, aged 60-80 years receiving optimal pharmacotherapy treatment (79% men, 18% diabetes). Methods Cystatin C based estimated glomerular filtration rate (eGFRcysC) was calculated at baseline and after 41 months, using the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation. Obesity was defined as body mass index ≥ 30 kg/m2 and high waist circumference as ≥102 and ≥88 cm for men and women. The relation between body mass index, waist circumference and annual eGFRcysC decline was evaluated by linear regression. Results At baseline, mean (standard deviation) eGFRcysC was 81.5 (19.6) ml/min/1.73 m2, 23% of all patients were obese. After multivariable adjustment, the annual mean (95% confidence interval) eGFRcysC decline in men and women was -1.45 (-1.59 to -1.31) and -0.92 (-1.20 to -0.63) ml/min/1.73 m2, respectively ( p = 0.001). Obese versus non-obese patients and patients with high versus normal waist circumference experienced greater annual eGFRcysC decline. Men and women showed an additional annual eGFRcysC decline of -0.35 (-0.56 to -0.14) and -0.21 (-0.55 to 0.14) ml/min/1.73 m2 per 5 kg/m2 body mass index increment ( p for interaction 0.3). Conclusions High compared to normal body mass index or waist circumference were associated with more rapid kidney function decline in older stable post-myocardial infarction patients receiving optimal drug therapy.
Collapse
Affiliation(s)
- Kevin Esmeijer
- Department of Nephrology, Leiden University Medical Center, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, The Netherlands
| | | | - Erik J Giltay
- Department of Psychiatry, Leiden University Medical Center, The Netherlands
| | - Theo Stijnen
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, The Netherlands
| | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, The Netherlands
| | - Johan W de Fijter
- Department of Nephrology, Leiden University Medical Center, The Netherlands
| | - Daan Kromhout
- Division of Human Nutrition, Wageningen University, The Netherlands
- Department of Epidemiology, University Medical Center Groningen, The Netherlands
| | - Ellen K Hoogeveen
- Department of Nephrology, Leiden University Medical Center, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, The Netherlands
- Department of Nephrology, Jeroen Bosch Hospital, The Netherlands
| |
Collapse
|
340
|
Titan SM, Pecoits-Filho R, Barreto SM, Lopes AA, Bensenor IJ, Lotufo PA. GlycA, a marker of protein glycosylation, is related to albuminuria and estimated glomerular filtration rate: the ELSA-Brasil study. BMC Nephrol 2017; 18:367. [PMID: 29262791 PMCID: PMC5738692 DOI: 10.1186/s12882-017-0779-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
Background Systemic inflammation has been implicated in several chronic diseases. GlycA is a new nuclear mass resonance (NMR) spectroscopy-derived biomarker of systemic inflammation that reflects protein glycosylation. We evaluated the association of GlycA with albuminuria and eGFR in the ELSA-Brasil Study. Methods The cross-sectional association between GlycA (automated NMR LipoProfile(®) test spectra, LabCorp, Raleigh, NC), and overnight 12 h–albuminuria and CKD-EPI eGFR was evaluated among 5050 participants. Results GlycA was higher among older, women, smokers, alcohol abstemious, obese and in those with diabetes, hypertension or dyslipidemia. In addition, both eGFR and albuminuria were associated to GlycA. In linear regression, GlycA was independently associated with log albuminuria (B 0.03; 95%CI 0.02–0.04, P < 0.0001, per 1sd increase) and inversely related to eGFR (B -0.53; 95%CI -0.99 – -0.07, P < 0.02), even after adjustments including hsCRP. In logistic regression, GlycA was independently related to the risk of A2 or A3 albuminuria (OR 1.42, 95%CI 1.27–1.57, p < 0.0001, per 1sd increase), of having an eGFR < 60 ml/min/1.73m2 (OR 1.26, 95%CI 1.12–1.41, p = 0.0003, per 1 sd) or of a combined diagnosis of both conditions (OR 1.35, 95%CI 1.23–1.46, p < 0.0001, per 1 sd). In the ROC curve, GlycA had a higher AUC in comparison to hsCRP (AUC 0.67 vs. 0.62, p = 0.06) for the association with albuminuria A2 or A3. Conclusions The present study demonstrates that GlycA is associated with albuminuria and eGFR, independently of major risk factors for CKD progression, including (and with a stronger association than) hsCRP. GlycA should be further evaluated in CKD progression.
Collapse
Affiliation(s)
- Silvia M Titan
- Nephrology Unit, Department of Medicine, Medical School, University of Sao Paulo, Av Dr Enéas de Carvalho Aguiar 255, Cerqueira César, São Paulo - SP, 05403-000, Brazil. .,Center for Clinical and Epidemiologic Research, Hospital Universitario, University of São Paulo, Av Prof. Lineu Prestes 2565, Butantã, São Paulo - SP, 05508-000, Brazil.
| | - Roberto Pecoits-Filho
- Department of Medicine, Medical School, Pontifícia Universidade Católica do Paraná, R. Imac. Conceição 1155. Prado Velho, Curitiba, PR, 80215-901, Brazil
| | - Sandhi M Barreto
- Department of Social and Preventive Medicine, Medical School, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena 190. Santa Efigênia, Belo Horizonte, MG, 30130-100, Brazil
| | - Antônio Alberto Lopes
- Clinical Epidemiology and Evidence Based-Medicine, University Hospital Professor Edgard Santos, Federal University of Bahia, Rua Augusto Viana, sn°. Canela, Salvador, BA, 40110-060, Brazil
| | - Isabela J Bensenor
- Center for Clinical and Epidemiologic Research, Hospital Universitario, University of São Paulo, Av Prof. Lineu Prestes 2565, Butantã, São Paulo - SP, 05508-000, Brazil.,General Medicine Unit, Department of Medicine, Medical School, University of Sao Paulo, Av Dr Enéas de Carvalho Aguiar 255, Cerqueira César, São Paulo - SP, 05403-000, Brazil
| | - Paulo A Lotufo
- Center for Clinical and Epidemiologic Research, Hospital Universitario, University of São Paulo, Av Prof. Lineu Prestes 2565, Butantã, São Paulo - SP, 05508-000, Brazil.,General Medicine Unit, Department of Medicine, Medical School, University of Sao Paulo, Av Dr Enéas de Carvalho Aguiar 255, Cerqueira César, São Paulo - SP, 05403-000, Brazil
| |
Collapse
|
341
|
Tian J, Niu L, An X. Cardiovascular risks in chronic kidney disease pediatric patients. Exp Ther Med 2017; 14:4615-4619. [PMID: 29201159 PMCID: PMC5704347 DOI: 10.3892/etm.2017.5117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/14/2017] [Indexed: 11/10/2022] Open
Abstract
One of the common factors for the premature death in children is advanced chronic kidney disease (CKD). Most often cardiovascular disease (CVD) is the reason for mortality. The cardiovascular (CV) morbidity starts early in the disease process and renal transplanted children (CKD-T) are also at risk. The present review is focused on the current views of the cardiovascular risks during CKD in pediatric patients. Variable data sources for the latest literature collection were explored which mainly included PubMed and Google Scholar. The most important risk factors for subclinical CVD were a young age, elevated BMI and systolic blood pressure z-scores as well as a low GFR and present albuminuria. Increasing blood pressure and BMI over follow-up were also important cardiac risk factors longitudinally. The present review concludes that altered cardiac function and remodeling are a concurrent part of the CKD process, start early in the disease development, and persist after renal transplantation. The findings suggest that children with CKD or CKD-T are at high risk for future CVD where younger patients with elevated BMI and slightly increased blood pressures, as well as present albuminuria, are those at greatest risk, thus indicating targets for future interventions.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Ling Niu
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Xinjiang An
- Department of Pediatric Internal Medicine, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
342
|
George G, Patel N, Jang C, Wheeler D, Yaddanapudi SS, Dissin J, Balu R, Rangaswami J. Proteinuria Predicts Resistance to Antiplatelet Therapy in Ischemic Stroke. Transl Stroke Res 2017; 9:130-134. [PMID: 28875481 DOI: 10.1007/s12975-017-0568-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022]
Abstract
The occurrence of a stroke while on antiplatelet agents presents a therapeutic dilemma. One of the main causes for recurrent strokes is antiplatelet resistance more commonly known as high on treatment platelet reactivity (HTPR). Prior studies have established that proteinuria is associated with HTPR following myocardial infarction. Here, we investigated whether dipstick proteinuria correlates with HTPR in patients presenting with stroke. We performed a retrospective cohort analysis of 102 patients admitted for a recurrent ischemic stroke that had either a VerifyNow aspirin or VerifyNow clopidogrel laboratory test performed to assess platelet reactivity. Dipstick proteinuria was defined as > 30 mg/dl (2+ or more). HTPR was defined as an aspirin resistance unit > 550 for aspirin and a P2Y12 reactivity unit > 208 for clopidogrel. Patients with proteinuria on dipstick were significantly more likely to have HTPR to either aspirin (p value 0.017) or clopidogrel (p value 0.017). After controlling for age, smoking, diabetes, hypertension, CAD and GFR, proteinuria was an independent predictor of HTPR for patient taking aspirin (p = 0.025). Platelet resistance is an entity that undermines the activity of antiplatelet agents in reducing stroke risk. Here, we demonstrate an association with increased platelet reactivity and proteinuria. This highlights a potential new therapeutic target in reducing future stroke risk.
Collapse
Affiliation(s)
- Gemlyn George
- Department of Internal Medicine, Einstein Medical Center, Klein 303, 5501 Old York Road, Philadelphia, PA, 19141, USA.
| | - Nikul Patel
- Department of Internal Medicine, Einstein Medical Center, Klein 303, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - Cecilia Jang
- Department of Internal Medicine, Einstein Medical Center, Klein 303, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - David Wheeler
- Department of Internal Medicine, Einstein Medical Center, Klein 303, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - Sridhara S Yaddanapudi
- Department of Neurosciences, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - Jonathan Dissin
- Department of Neurosciences, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - Ramani Balu
- Division of Neurocritical Care, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Janani Rangaswami
- Department of Internal Medicine, Einstein Medical Center, Klein 303, 5501 Old York Road, Philadelphia, PA, 19141, USA
- Sidney Kimmel College of Thomas Jefferson University, Philadelphia, PA, USA
- Delaware Valley Nephrology Associates, Philadelphia, PA, USA
| |
Collapse
|
343
|
Kraut JA, Madias NE. Adverse Effects of the Metabolic Acidosis of Chronic Kidney Disease. Adv Chronic Kidney Dis 2017; 24:289-297. [PMID: 29031355 DOI: 10.1053/j.ackd.2017.06.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/27/2017] [Indexed: 01/01/2023]
Abstract
The kidney has the principal role in the maintenance of acid-base balance, and therefore, a fall in renal net acid excretion and positive H+ balance often leading to reduced serum [HCO3-] are observed in the course of CKD. This metabolic acidosis can be associated with muscle wasting, development or exacerbation of bone disease, hypoalbuminemia, increased inflammation, progression of CKD, protein malnutrition, alterations in insulin, leptin, and growth hormone, and increased mortality. Importantly, some of the adverse effects can be observed even in the absence of overt hypobicarbonatemia. Administration of base decreases muscle wasting, improves bone disease, restores responsiveness to insulin, slows progression of CKD, and possibly reduces mortality. Base is recommended when serum [HCO3-] is <22 mEq/L, but the target serum [HCO3-] remains unclear. Evidence that increments of serum [HCO3-] >26 mEq/L might be associated with worsening of cardiovascular disease adds complexity to treatment decisions. Further study of the mechanisms through which positive H+ balance in CKD contributes to its various adverse effects and the pathways involved in mediating the benefits and complications of base therapy is warranted.
Collapse
Affiliation(s)
- Jeffrey A Kraut
- Medical and Research Services VHAGLA Healthcare System, UCLA Membrane Biology Laboratory and Division of Nephrology VHAGLA Healthcare System and David Geffen School of Medicine, Los Angeles, CA; and Division of Nephrology, Department of Medicine, St. Elizabeth's Medical Center and Department of Medicine, Tufts University School of Medicine, Boston, MA
| | - Nicolaos E Madias
- Medical and Research Services VHAGLA Healthcare System, UCLA Membrane Biology Laboratory and Division of Nephrology VHAGLA Healthcare System and David Geffen School of Medicine, Los Angeles, CA; and Division of Nephrology, Department of Medicine, St. Elizabeth's Medical Center and Department of Medicine, Tufts University School of Medicine, Boston, MA.
| |
Collapse
|
344
|
Shih CP, Lin HC, Chung CH, Hsiao PJ, Wang CH, Lee JC, Chien WC. Increased risk of tinnitus in patients with chronic kidney disease: A nationwide, population-based cohort study. PLoS One 2017; 12:e0183192. [PMID: 28813508 PMCID: PMC5557597 DOI: 10.1371/journal.pone.0183192] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/31/2017] [Indexed: 12/27/2022] Open
Abstract
Tinnitus mostly results from central and peripheral auditory pathology. Chronic kidney disease (CKD) is a major risk factor for cerebrovascular disease. However, no studies have evaluated the association between tinnitus and CKD. The aim of this study is to investigate the risk of tinnitus in patients with CKD. This retrospective cohort study was conducted using Taiwan National Health Insurance Research Database from 2000 to 2010. We established a CKD group (n = 185,430) and a non-CKD comparison group (n = 556,290) to investigate the incidence of tinnitus. Cox proportional hazard regression analysis was used to evaluate the effects of CKD on tinnitus risk. The results showed CKD significantly increased the risk of tinnitus (adjusted hazard ratio, 3.02; 95% CI, 2.655-3.456, P<0.001). A subgroup analysis revealed the increase in risk of tinnitus is more in CKD patients with heart failure (adjusted hazard ratio, 9.975; 95% CI, 5.001-18.752) and diabetes mellitus (adjusted hazard ratio, 3.712; 95% CI, 2.856-5.007). Furthermore, compared to non-CKD patients, the risk of tinnitus was increased 4.586-fold (95% CI, 2.399-6.7) in CKD patients with dialysis and 2.461-fold (95% CI, 1.033-3.454) in CKD patients without dialysis. This study is the first to report that CKD is associated with an increased risk of tinnitus. Among CKD cohort, patients with dialysis are at a higher risk of tinnitus than those without dialysis.
Collapse
Affiliation(s)
- Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hung-Che Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chi-Hsiang Chung
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Medical Research, Tri-Service General Hospital, Taipei, Taiwan, ROC
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan, ROC
| | - Chih-Hung Wang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Jih-Chin Lee
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Wu-Chien Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Medical Research, Tri-Service General Hospital, Taipei, Taiwan, ROC
- * E-mail:
| |
Collapse
|
345
|
Choi JW, Oh IH, Lee CH, Park JS. Is there a J-shaped relationship between the fatty liver index and risk of microalbuminuria in the general population? Clin Chim Acta 2017; 481:231-237. [PMID: 28818597 DOI: 10.1016/j.cca.2017.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/11/2017] [Accepted: 08/13/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND We investigated whether the fatty liver index (FLI), as a clinical indicator of hepatic fat accumulation based on body mass index, gamma-glutamyl-transferase, triglycerides, and waist circumference, has an association with microalbuminuria. METHODS We analyzed anthropometric and biochemical data from a nation-wide, population-based, cross-sectional study. A total of 1605 participants included were healthy native Korean 40years or older and divided into quintiles according to their log-FLI and sex. Microalbuminuria was defined as urine albumin/creatinine ratio (UACR) between 30 and 300mg/g. RESULTS Participants in higher quintiles of log-FLI were more obese and hypertensive and had greater glycemic exposure, poorer lipid profiles, and greater increases in log-UACR compared with lower quintiles. Linear regression analysis demonstrated that log-FLI was associated with systolic and diastolic blood pressure, body mass index, waist circumference, fasting plasma glucose, glycated hemoglobin, and log-UACR. In logistic regression adjusted for age, sex, body mass index, waist circumference, and fasting plasma glucose, the OR of microalbuminuria was elevated in quintile 1 (adjusted OR=2.161, 95% CI=0.453-10.31) and quintile 5 (adjusted OR=6.387, 95% CI=1.317-51.58), when compared to quintile 2. CONCLUSIONS There appears to be a J-shaped association between FLI and UACR in healthy general population.
Collapse
Affiliation(s)
- Jong Wook Choi
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Il Hwan Oh
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Chang Hwa Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea.
| | - Joon-Sung Park
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
346
|
Netea MG, Balkwill F, Chonchol M, Cominelli F, Donath MY, Giamarellos-Bourboulis EJ, Golenbock D, Gresnigt MS, Heneka MT, Hoffman HM, Hotchkiss R, Joosten LA, Kastner DL, Korte M, Latz E, Libby P, Mandrup-Poulsen T, Mantovani A, Mills KHG, Nowak KL, O’Neill LA, Pickkers P, van der Poll T, Ridker PM, Schalkwijk J, Schwartz DA, Siegmund B, Steer CJ, Tilg H, van der Meer JW, van de Veerdonk FL, Dinarello CA. A guiding map for inflammation. Nat Immunol 2017; 18:826-831. [PMID: 28722720 PMCID: PMC5939996 DOI: 10.1038/ni.3790] [Citation(s) in RCA: 521] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Biologists, physicians and immunologists have contributed to the understanding of the cellular participants and biological pathways involved in inflammation. Here, we provide a general guide to the cellular and humoral contributors to inflammation as well as to the pathways that characterize inflammation in specific organs and tissues.
Collapse
Affiliation(s)
- Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Frances Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism University Hospital and University of Basel, Switzerland
| | | | - Douglas Golenbock
- Division of Infectious Diseases and Immunology, University of Massacchussetts Medical School, Worchester, USA
| | - Mark S. Gresnigt
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michael T. Heneka
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn, Bonn, Germany
| | - Hal M. Hoffman
- Division of Pediatric Allergy, Immunology, and Rheumatology, University of California at San Diego and Rady Children’s Hospital of San Diego, USA
| | - Richard Hotchkiss
- Department of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca Romania
| | - Daniel L. Kastner
- Inflammatory Disease Section, Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Martin Korte
- TU Braunschweig, Zoological Institute, Braunschweig, Germany and HZI, AG NIND, Braunschweig, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
- Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA, USA
| | - Peter Libby
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Alberto Mantovani
- Humanitas University and Humanitas Clinica Research Center, Rozzano, Milano, Italy
| | - Kingston H. G. Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Kristen L. Nowak
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, USA
| | - Luke A. O’Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul M. Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Joost Schalkwijk
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David A. Schwartz
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, USA
| | - Britta Siegmund
- Department of Medicine (Gastroenterology, Infectious Diseases, Rheumatology), Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Clifford J. Steer
- Departments of Medicine and Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology, Medical University Innsbruck, Austria
| | - Jos W.M. van der Meer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charles A. Dinarello
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
347
|
Miranda A, Cordeiro T, dos Santos Lacerda Soares TM, Ferreira R, Simões e Silva A. Kidney–brain axis inflammatory cross-talk: from bench to bedside. Clin Sci (Lond) 2017; 131:1093-1105. [DOI: 10.1042/cs20160927] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Epidemiologic data suggest that individuals at all stages of chronic kidney disease (CKD) have a higher risk of developing neuropsychiatric disorders, cognitive impairment, and dementia. This risk is generally explained by the high prevalence of both symptomatic and subclinical ischemic cerebrovascular lesions. However, other potential mechanisms, including cytokine/chemokine release, production of reactive oxygen species (ROS), circulating and local formation of trophic factors and of renin–angiotensin system (RAS) molecules, could also be involved, especially in the absence of obvious cerebrovascular disease. In this review, we discuss experimental and clinical evidence for the role of these mechanisms in kidney–brain cross-talk. In addition, we hypothesize potential pathways for the interactions between kidney and brain and their pathophysiological role in neuropsychiatric and cognitive changes found in patients with CKD. Understanding the pathophysiologic interactions between renal impairment and brain function is important in order to minimize the risk for future cognitive impairment and to develop new strategies for innovative pharmacological treatment.
Collapse
Affiliation(s)
- Aline Silva Miranda
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, UFMG, Belo Horizonte, Brazil
| | - Thiago Macedo Cordeiro
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, UFMG, Belo Horizonte, Brazil
| | | | - Rodrigo Novaes Ferreira
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Ana Cristina Simões e Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, UFMG, Belo Horizonte, Brazil
| |
Collapse
|
348
|
Boucquemont J, Lawson-Ayayi S, Rigothier C, Bonnet F, Proust-Lima C, Neau D, Greib C, Miremont-Salamé G, Dabis F, Dupon M, Dauchy FA. Absence of Decline of Kidney Function in Human Immunodeficiency Virus-Infected Patients Under Routine Clinical Management. Nephron Clin Pract 2017; 136:211-220. [PMID: 28445881 DOI: 10.1159/000467400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/27/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Since the introduction of antiretroviral therapy (ART), human immunodeficiency virus (HIV)-infected patients have a drastically improved prognosis but at the same time they are also more affected by non-HIV related complications, such as chronic kidney disease. The objective of our study was to investigate the effect of proteinuria and tenofovir (TDF)-containing ART regimens on the temporal evolution of estimated glomerular filtration rate (eGFR). METHODS Between April 2008 and October 2012, we enrolled 395 patients with a complete renal evaluation among patients from the ANRS C03 Aquitaine cohort, a prospective hospital-based cohort of HIV-1-infected patients under routine clinical management in southwestern France. eGFR was estimated at each patient follow-up visit. A linear mixed model was used to analyze eGFR dynamics, accounting for change in TDF by modeling eGFR trajectory according to treatment periods. RESULTS At inclusion, 56.7% of patients were treated with TDF-containing ART regimens; prevalence of glomerular and tubular proteinuria was 7.9 and 10.8% respectively. A 1-year increase of cumulative exposure to TDF was significantly associated with a mean eGFR decrease of 1.27 mL/min/1.73 m2 (95% CI [-2.14 to -0.41]). Only a urine protein to creatinine ratio >100 mg/mmol and/or a urine albumin to creatinine ratio >70 mg/mmol were associated with eGFR trajectory (mean slope 6.18 mL/min/1.73 m2 per year; 95% CI [2.71 to 9.65]), whereas TDF use was not associated with such eGFR temporal evolution. CONCLUSION Decline in kidney function is limited under routine clinical management with monitoring of renal function and interventions including decision to continue or discontinue TDF.
Collapse
Affiliation(s)
- Julie Boucquemont
- INSERM U1219 Bordeaux Population Health, ISPED, University of Bordeaux, Bordeaux, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
349
|
Zoccali C, Vanholder R, Massy ZA, Ortiz A, Sarafidis P, Dekker FW, Fliser D, Fouque D, Heine GH, Jager KJ, Kanbay M, Mallamaci F, Parati G, Rossignol P, Wiecek A, London G. The systemic nature of CKD. Nat Rev Nephrol 2017; 13:344-358. [PMID: 28435157 DOI: 10.1038/nrneph.2017.52] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The accurate definition and staging of chronic kidney disease (CKD) is one of the major achievements of modern nephrology. Intensive research is now being undertaken to unravel the risk factors and pathophysiologic underpinnings of this disease. In particular, the relationships between the kidney and other organs have been comprehensively investigated in experimental and clinical studies in the last two decades. Owing to technological and analytical limitations, these links have been studied with a reductionist approach focusing on two organs at a time, such as the heart and the kidney or the bone and the kidney. Here, we discuss studies that highlight the complex and systemic nature of CKD. Energy balance, innate immunity and neuroendocrine signalling are highly integrated biological phenomena. The diseased kidney disrupts such integration and generates a high-risk phenotype with a clinical profile encompassing inflammation, protein-energy wasting, altered function of the autonomic and central nervous systems and cardiopulmonary, vascular and bone diseases. A systems biology approach to CKD using omics techniques will hopefully enable in-depth study of the pathophysiology of this systemic disease, and has the potential to unravel critical pathways that can be targeted for CKD prevention and therapy.
Collapse
Affiliation(s)
- Carmine Zoccali
- CNR-IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension Unit, Ospedali Riuniti 89124 Reggio Calabria, Italy
| | - Raymond Vanholder
- Ghent University Hospital, Department of Nephrology, Department of Internal Medicine, University Hospital Gent, De Pintelaan 185, B9000 Ghent, Belgium
| | - Ziad A Massy
- Division of Nephrology, Ambroise Paré Hospital, Assistance Publique Hôpitaux de Paris, 9 Avenue Charles de Gaulle, 92100 Boulogne-Billancourt, Paris.,University of Paris Ouest-Versailles-Saint-Quentin-en-Yvelines (UVSQ), 55 Avenue de Paris, 78000 Versailles, France.,Inserm U-1018, Centre de recherche en épidémiologie et santé des populations (CESP), Equipe 5, Hôpital Paul-Brousse, 16 avenue Paul Vaillant-Couturier, 94807 Villejuif Cedex, France.,Paris-Sud University (PSU), 15 Rue Georges Clemenceau, 91400 Orsay, France.,French-Clinical Research Infrastructure Network (F-CRIN), Pavillon Leriche 2è étage CHU de Toulouse, Place Dr Baylac TSA40031, 31059 TOULOUSE Cedex 3, France
| | - Alberto Ortiz
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Av. Reyes Católicos, 2, 28040 Madrid, Spain
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Thessaloniki, Konstantinoupoleos 49, Thessaloniki 546 42, Greece
| | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Danilo Fliser
- Department Internal Medicine IV-Renal and Hypertensive Disease-Saarland University Medical Centre Kirrberger Straß 66421 Homburg, Saar, Germany
| | - Denis Fouque
- Université de Lyon, UCBL, Carmen, Department of Nephrology, Centre Hospitalier Lyon-Sud, F-69495 Pierre Bénite, France
| | - Gunnar H Heine
- Department Internal Medicine IV-Renal and Hypertensive Disease-Saarland University Medical Centre Kirrberger Straß 66421 Homburg, Saar, Germany
| | - Kitty J Jager
- European Renal Association-European Dialysis and Transplant Association (ERA-EDTA) Registry, Department of Medical Informatics, Meibergdreef 9, 1105 AZ Amsterdam-Zuidoost, The Netherlands
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine,Koç University, Rumelifeneri Yolu 34450 Sarıyer Istanbul, Turkey
| | - Francesca Mallamaci
- CNR-IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension Unit, Ospedali Riuniti 89124 Reggio Calabria, Italy.,Nephrology, Dialysis and Transplantation Unit Ospedali Riuniti, 89124 Reggio Calabria Italy
| | - Gianfranco Parati
- Department of Cardiovascular, Neural and Metabolic Sciences, S. Luca Hospital, Istituto Auxologico Italiano &Department of Medicine and Surgery, University of Milan-Bicocca, Piazzale Brescia 20, Milan 20149, Italy
| | - Patrick Rossignol
- French-Clinical Research Infrastructure Network (F-CRIN), Pavillon Leriche 2è étage CHU de Toulouse, Place Dr Baylac TSA40031, 31059 TOULOUSE Cedex 3, France.,Inserm, Centre d'Investigations Cliniques-Plurithématique 1433, Cardiovascular and Renal Clinical Trialists (INI-CRCT), Institut Lorrain du Cœur et des Vaisseaux Louis Mathieu, 4 rue Morvan, 54500 Vandoeuvre-les-Nancy, France.,Inserm U1116, Faculté de Médecine, Bâtiment D 1er étage, 9 avenue de la forêt de Haye - BP 184, 54500 Vandœuvre-lès-Nancy Cedex, France.,CHU Nancy, Département de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, 5 Rue du Morvan, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 34 Cours Léopold, 54000 Nancy, France
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Francuska 20/24 Street, Pl-40-027 Katowice, Poland
| | - Gerard London
- INSERM U970, Hopital Européen Georges Pompidou, 20 Rue Leblanc, 75015 Paris, France
| | | |
Collapse
|
350
|
Yang SK, Liu J, Yi B, Mao J, Zhang XM, Liu Y, Lei DD, Gui M, Zhang H. Elevated High Sensitivity C-Reactive Protein Increases the Risk of Microalbuminuria in Subjects With Cardiovascular Disease Risk Factors. Ther Apher Dial 2017; 21:387-394. [PMID: 28322017 DOI: 10.1111/1744-9987.12530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 12/26/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022]
Abstract
Microalbuminuria is an early biomarker of kidney injury, the aim of this study was to investigate the impact of high sensitivity C-reactive protein (hs-CRP) on microalbuminuria in adults with cardiovascular disease (CVD) risk factors. A total of 5667 subjects who voluntarily attended annual health screenings in the Third Xiangya Hospital of Central South University were included in the study. Logistic regression was used to determine the validity of hs-CRP to predict the presence of microalbuminuria. In the unadjusted regression analysis, male gender (OR 2.23), age (OR 1.71), smoking status (OR 1.48), obesity (OR 2.41), hypertension(OR 4.03), diabetes (OR 4.61), hyperuricemia (OR 1.83), and high hs-CRP(OR 1.61) were associated with microalbuminuria. Multivariate logistic regression analysis showed that the adjusted odds ratios (OR) of diabetes, hypertension, male, abdominal obesity, and high hs-CRP for microalbuminuria were 2.71, 2.57, 1.51, 1.28 and 1.41 in all subjects, respectively (P < 0.01). ROC analysis indicated that the cut-off for hs-CRP with better properties for screening of microalbuminuria was identified as ≥0.85 mg/L. The ORs of microalbuminuria were 1.97, 1.49, 1.32 and 1.71 times in male, abdominal obesity, diabetes and hypertension subjects with hs-CRP ≥ 0.85 mg/L, respectively, compared with those without elevated hs-CRP (P < 0.01). Elevated hs-CRP level was associated with microalbuminuria independent of cardiovascular risk factors. In addition, elevated hs-CRP increased the risk of developing microalbuminuria in subjects with CVD risk factors, indicating that chronic inflammation could amplify the deterioration of kidney by effect of CVD risk factors.
Collapse
Affiliation(s)
- Shi-Kun Yang
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Liu
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bin Yi
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Mao
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Ming Zhang
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan-Dan Lei
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Gui
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|