ijms-logo

Journal Browser

Journal Browser

Advances in Chronic Kidney Disease 2017

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (31 August 2017) | Viewed by 118419

Special Issue Editor


E-Mail Website
Guest Editor
Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
Interests: aging; acute kidney injury; cell adhesion; nephrotoxicity
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The prevalence of chronic kidney disease (CKD) continues to expand worldwide, and the morbidity and health care costs associated with CKD impacts health care costs across the globe. While much effort has gone into understanding the pathogenesis of CKD, progress on therapeutic options to prevent, or attenuate, the progression of renal dysfunction has been much slower.

This issue of the International Journal of Molecular Sciences will focus on recent “Advances in Chronic Kidney Disease”, including new insights into the epidemiology, pathogenesis, and treatment of CKD. While estimated glomerular filtration rate (GFR) and proteinuria are widely used markers of CKD progression, new data is emerging on other potential biomarkers for the disease. In addition, several other morbidities, including acute kidney injury (AKI), are associated with CKD and submissions dealing with these diseases are welcome.

Assoc. Prof. Dr. Alan Parrish
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • chronic kidney disease
  • acute kidney injury
  • fibrosis
  • inflammation
  • aging
  • diabetes
  • hypertension
  • biomarkers
  • proteinuria
  • therapeutics

Published Papers (16 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

12 pages, 902 KiB  
Article
A Simple Method to Measure Renal Function in Swine by the Plasma Clearance of Iohexol
by Sergio Luis-Lima, Consolación García-Contreras, Marta Vázquez-Gómez, Susana Astiz, Fabiola Carrara, Flavio Gaspari, Natalia Negrín-Mena, Alejandro Jiménez-Sosa, Hugo Jiménez-Hernández, Antonio González-Bulnes and Esteban Porrini
Int. J. Mol. Sci. 2018, 19(1), 232; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19010232 - 12 Jan 2018
Cited by 10 | Viewed by 4527
Abstract
There is no simple method to measure glomerular filtration rate (GFR) in swine, an established model for studying renal disease. We developed a protocol to measure GFR in conscious swine by using the plasma clearance of iohexol. We used two groups, test and [...] Read more.
There is no simple method to measure glomerular filtration rate (GFR) in swine, an established model for studying renal disease. We developed a protocol to measure GFR in conscious swine by using the plasma clearance of iohexol. We used two groups, test and validation, with eight animals each. Ten milliliters of iohexol (6.47 g) was injected into the marginal auricular vein and blood samples (3 mL) were collected from the orbital sinus at different points after injection. GFR was determined using two models: two-compartment (CL2: all samples) and one-compartment (CL1: the last six samples). In the test group, CL1 overestimated CL2 by ~30%: CL2 = 245 ± 93 and CL1 = 308 ± 123 mL/min. This error was corrected by a first-order polynomial quadratic equation to CL1, which was considered the simplified method: SM = −47.909 + (1.176xCL1) − (0.00063968xCL12). The SM showed narrow limits of agreement with CL2, a concordance correlation of 0.97, and a total deviation index of 14.73%. Similar results were obtained for the validation group. This protocol is reliable, reproducible, can be performed in conscious animals, uses a single dose of the marker, and requires a reduced number of samples, and avoids urine collection. Finally, it presents a significant improvement in animal welfare conditions and handling necessities in experimental trials. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

1662 KiB  
Article
Rho-Kinase Blockade Attenuates Podocyte Apoptosis by Inhibiting the Notch Signaling Pathway in Diabetic Nephropathy
by Keiichiro Matoba, Daiji Kawanami, Yosuke Nagai, Yusuke Takeda, Tomoyo Akamine, Sho Ishizawa, Yasushi Kanazawa, Tamotsu Yokota and Kazunori Utsunomiya
Int. J. Mol. Sci. 2017, 18(8), 1795; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081795 - 18 Aug 2017
Cited by 33 | Viewed by 5711
Abstract
Podocyte apoptosis is a key process in the onset of diabetic nephropathy. A significant body of evidence shows that the Notch signaling pathway plays a central role in this process. We found that Rho-kinase mediates transforming growth factor β (TGF-β)-induced Notch ligand Jag1 [...] Read more.
Podocyte apoptosis is a key process in the onset of diabetic nephropathy. A significant body of evidence shows that the Notch signaling pathway plays a central role in this process. We found that Rho-kinase mediates transforming growth factor β (TGF-β)-induced Notch ligand Jag1 expression. Importantly, TGF-β-mediated podocyte apoptosis was attenuated by Rho-kinase inhibition. Mechanistically, Rho-kinase regulated Jag1 induction via the extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK) but not Smad pathways. Consistently, the Rho-kinase inhibitor fasudil prevented albuminuria and the urinary excretion of nephrin in db/db mice and reduced the prevalence of podocyte apoptosis and Jag1 expression. Finally, the expression of Jag1 and apoptosis markers such as Bax and cyclin-dependent kinase inhibitor 1A (CDKN1A) was decreased in podocytes derived from db/db mice treated with fasudil. The present study provides evidence that Rho-kinase plays a key role in podocyte apoptosis. Rho-kinase is an attractive therapeutic target for diabetic nephropathy. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

3177 KiB  
Article
Endocytosis of Albumin Induces Matrix Metalloproteinase-9 by Activating the ERK Signaling Pathway in Renal Tubule Epithelial Cells
by Xiaoming Chen, Alyssa Cobbs, Jasmine George, Ashmeer Chima, Fidele Tuyishime and Xueying Zhao
Int. J. Mol. Sci. 2017, 18(8), 1758; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081758 - 12 Aug 2017
Cited by 9 | Viewed by 4848
Abstract
Matrix metalloproteinase-9 (MMP-9) is dysregulated in chronic kidney diseases including diabetic nephropathy. This study was performed to examine the expression of MMP-9 in renal tubule epithelial cells (TECs) under diabetic conditions and its regulatory mechanisms. We characterized MMP-9 protein in diabetic animals and [...] Read more.
Matrix metalloproteinase-9 (MMP-9) is dysregulated in chronic kidney diseases including diabetic nephropathy. This study was performed to examine the expression of MMP-9 in renal tubule epithelial cells (TECs) under diabetic conditions and its regulatory mechanisms. We characterized MMP-9 protein in diabetic animals and primary cultured rat TECs exposed to exogenous albumin and high glucose. We also used specific inhibitors to determine if internalization of albumin and/or extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation were required for MMP-9 secretion. Immunostaining of kidney sections revealed enhanced MMP-9 signal in the damaged proximal tubules in Zucker diabetic fatty (ZDF) rats. ZDF rats also exhibited an albuminuria-related and age-dependent increase in MMP-9 excretion, which was prevented by rosiglitazone. In primary cultured rat TECs, high glucose exposure did not increase MMP-9 secretion. In contrast, administration of rat serum albumin (RSA, 0.1–0.5 mg/mL) resulted in a dose-dependent increase in MMP-9 expression and secretion by TECs, which was abolished in the presence of an ERK1/2-specific inhibitor, U0126. Simvastatin, an inhibitor of albumin endocytosis, also prevented MMP-9 secretion. Taken together, these results demonstrate that endocytosis of albumin stimulates MMP-9 secretion by TECs through the ERK signaling pathway. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

1673 KiB  
Article
Plasma Neutrophil Gelatinase-Associated Lipocalin and Predicting Clinically Relevant Worsening Renal Function in Acute Heart Failure
by Kevin Damman, Mattia A. E. Valente, Dirk J. Van Veldhuisen, John G. F. Cleland, Christopher M. O’Connor, Marco Metra, Piotr Ponikowski, Gad Cotter, Beth Davison, Michael M. Givertz, Daniel M. Bloomfield, Hans L. Hillege and Adriaan A. Voors
Int. J. Mol. Sci. 2017, 18(7), 1470; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18071470 - 08 Jul 2017
Cited by 20 | Viewed by 4734
Abstract
The aim of this study was to evaluate the ability of Neutrophil Gelatinase-Associated Lipocalin (NGAL) to predict clinically relevant worsening renal function (WRF) in acute heart failure (AHF). Plasma NGAL and serum creatinine changes during the first 4 days of admission were investigated [...] Read more.
The aim of this study was to evaluate the ability of Neutrophil Gelatinase-Associated Lipocalin (NGAL) to predict clinically relevant worsening renal function (WRF) in acute heart failure (AHF). Plasma NGAL and serum creatinine changes during the first 4 days of admission were investigated in 1447 patients hospitalized for AHF and enrolled in the Placebo-Controlled Randomized Study of the Selective A1Adenosine Receptor Antagonist Rolofylline for Patients Hospitalized with Acute Decompensated Heart Failure and Volume Overload to Assess Treatment Effect on Congestion and Renal Function (PROTECT) study. WRF was defined as serum creatinine rise ≥ 0.3 mg/dL through day 4. Biomarker patterns were described using linear mixed models. WRF developed in 325 patients (22%). Plasma NGAL did not rise earlier than creatinine in patients with WRF. After multivariable adjustment, baseline plasma NGAL, but not creatinine, predicted WRF. AUCs for WRF prediction were modest (<0.60) for all models. NGAL did not independently predict death or rehospitalization (p = n.s.). Patients with WRF and high baseline plasma NGAL had a greater risk of death, and renal or cardiovascular rehospitalization by 60 days than patients with WRF and a low baseline plasma NGAL (p for interaction = 0.024). A rise in plasma NGAL after baseline was associated with a worse outcome in patients with WRF, but not in patients without WRF (p = 0.007). On the basis of these results, plasma NGAL does not provide additional, clinically relevant information about the occurrence of WRF in patients with AHF. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

1841 KiB  
Article
Urinary Angiotensinogen Could Be a Prognostic Marker of the Renoprotection of Olmesartan in Metabolic Syndrome Patients
by Tomoko Mizushige, Hiroyuki Kobori, Hirofumi Hitomi, Yoko Nishijima, Fumihiro Tomoda, Satoshi Morimoto, Masakazu Kohno and Akira Nishiyama
Int. J. Mol. Sci. 2016, 17(11), 1800; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17111800 - 27 Oct 2016
Cited by 2 | Viewed by 4315
Abstract
This study was performed to demonstrate urinary angiotensinogen as a potential prognostic marker of the albuminuria reduction effects of olmesartan in patients with metabolic syndrome. In 24 patients (eight women, 57.88 ± 2.00 years), 5–40 mg/day of olmesartan were given. Urinary concentrations of [...] Read more.
This study was performed to demonstrate urinary angiotensinogen as a potential prognostic marker of the albuminuria reduction effects of olmesartan in patients with metabolic syndrome. In 24 patients (eight women, 57.88 ± 2.00 years), 5–40 mg/day of olmesartan were given. Urinary concentrations of albumin and angiotensinogen (normalized by urinary concentrations of creatinine) and plasma renin activity were measured before and after the 12- and 24-week marks of olmesartan treatment. Olmesartan treatment increased plasma renin activity and decreased urinary albumin and urinary angiotensinogen significantly (p < 0.05). Based on the % change in urinary albumin, patients were divided into two groups, responders (<−50%) and non-responders (≥−50%), and a logistic analysis of urinary angiotensinogen before treatment showed the area under the curve as 0.694. When the cutoff value of urinary angiotensinogen before the treatment of 13.9 µg/g Cr was used, the maximum Youden index (0.500, specificity: 11/12 = 91.7% and sensitivity: 7/12 = 58.3%) was obtained. When all patients were re-divided into two groups, those with higher values of urinary angiotensinogen before the treatment (Group H, n = 16) and those with lower values, Group H showed significantly decreased urinary albumin (p < 0.05). Therefore, urinary angiotensinogen could be a prognostic marker of the albuminuria reduction effects of olmesartan in patients with metabolic syndrome. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

Review

Jump to: Research

1973 KiB  
Review
Xanthine Oxidase Inhibitors for Improving Renal Function in Chronic Kidney Disease Patients: An Updated Systematic Review and Meta-Analysis
by Anna Pisano, Valeria Cernaro, Guido Gembillo, Graziella D’Arrigo, Michele Buemi and Davide Bolignano
Int. J. Mol. Sci. 2017, 18(11), 2283; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18112283 - 31 Oct 2017
Cited by 44 | Viewed by 6267
Abstract
Background: Accruing evidence suggests that Xanthine Oxidase inhibitors (XOis) may bring direct renal benefits, besides those related to their hypo-uricemic effect. We hence aimed at performing a systematic review of randomized controlled trials (RCTs) to verify if treatment with XOis may improve renal [...] Read more.
Background: Accruing evidence suggests that Xanthine Oxidase inhibitors (XOis) may bring direct renal benefits, besides those related to their hypo-uricemic effect. We hence aimed at performing a systematic review of randomized controlled trials (RCTs) to verify if treatment with XOis may improve renal outcomes in individuals with chronic kidney disease (CKD). Methods: Ovid-MEDLINE, PubMed and CENTRAL databases were searched for RCTs comparing any XOi to standard therapy or placebo. The primary endpoint of interest was progression to End-Stage Kidney Disease (ESKD); secondary endpoints were changes in serum creatinine, glomerular filtration rate (eGFR), proteinuria and albuminuria. Results: XOis treatment significantly reduced the risk of ESKD compared to the control (3 studies, 204 pts; RR = 0.42; 95% CI, 0.22, 0.80) and also improved eGFR in data pooled from RCTs with long follow-up times (>3 mo.) (4 studies, 357 pts; mean difference (MD) 6.82 mL/min/1.73 m2; 95% CI, 3.50, 10.15) and high methodological quality (blind design) (3 studies, 400 pts; MD 2.61 mL/min/1.73 m2; 95% CI, 0.23, 4.99). Conversely, no definite effects were apparently noticed on serum creatinine, proteinuria and albuminuria. Conclusions: XOis may represent a promising tool for retarding disease progression in CKD patients. Future trials are awaited to confirm the generalizability of these findings to the whole CKD population. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

2333 KiB  
Review
Biomarkers for Chronic Kidney Disease Associated with High Salt Intake
by Keiko Hosohata
Int. J. Mol. Sci. 2017, 18(10), 2080; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18102080 - 30 Sep 2017
Cited by 28 | Viewed by 5922
Abstract
High salt intake has been related to the development to chronic kidney disease (CKD) as well as hypertension. In its early stages, symptoms of CKD are usually not apparent, especially those that are induced in a “silent” manner in normotensive individuals, thereby providing [...] Read more.
High salt intake has been related to the development to chronic kidney disease (CKD) as well as hypertension. In its early stages, symptoms of CKD are usually not apparent, especially those that are induced in a “silent” manner in normotensive individuals, thereby providing a need for some kind of urinary biomarker to detect injury at an early stage. Because traditional renal biomarkers such as serum creatinine are insensitive, it is difficult to detect kidney injury induced by a high-salt diet, especially in normotensive individuals. Recently, several new biomarkers for damage of renal tubular epithelia such as neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (Kim-1) have been identified. Previously, we found a novel renal biomarker, urinary vanin-1, in several animal models with renal tubular injury. However, there are few studies about early biomarkers of the progression to CKD associated with a high-salt diet. This review presents some new insights about these novel biomarkers for CKD in normotensives and hypertensives under a high salt intake. Interestingly, our recent reports using spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto rats (WKY) fed a high-salt diet revealed that urinary vanin-1 and NGAL are earlier biomarkers of renal tubular damage in SHR and WKY, whereas urinary Kim-1 is only useful as a biomarker of salt-induced renal injury in SHR. Clinical studies will be needed to clarify these findings. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

904 KiB  
Review
Novel Biomarkers in the Diagnosis of Chronic Kidney Disease and the Prediction of Its Outcome
by Jacek Rysz, Anna Gluba-Brzózka, Beata Franczyk, Zbigniew Jabłonowski and Aleksandra Ciałkowska-Rysz
Int. J. Mol. Sci. 2017, 18(8), 1702; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081702 - 04 Aug 2017
Cited by 188 | Viewed by 13371
Abstract
In its early stages, symptoms of chronic kidney disease (CKD) are usually not apparent. Significant reduction of the kidney function is the first obvious sign of disease. If diagnosed early (stages 1 to 3), the progression of CKD can be altered and complications [...] Read more.
In its early stages, symptoms of chronic kidney disease (CKD) are usually not apparent. Significant reduction of the kidney function is the first obvious sign of disease. If diagnosed early (stages 1 to 3), the progression of CKD can be altered and complications reduced. In stages 4 and 5 extensive kidney damage is observed, which usually results in end-stage renal failure. Currently, the diagnosis of CKD is made usually on the levels of blood urea and serum creatinine (sCr), however, sCr has been shown to be lacking high predictive value. Due to the development of genomics, epigenetics, transcriptomics, proteomics, and metabolomics, the introduction of novel techniques will allow for the identification of novel biomarkers in renal diseases. This review presents some new possible biomarkers in the diagnosis of CKD and in the prediction of outcome, including asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA), uromodulin, kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), miRNA, ncRNA, and lincRNA biomarkers and proteomic and metabolomic biomarkers. Complicated pathomechanisms of CKD development and progression require not a single marker but their combination in order to mirror all types of alterations occurring in the course of this disease. It seems that in the not so distant future, conventional markers may be exchanged for new ones, however, confirmation of their efficacy, sensitivity and specificity as well as the reduction of analysis costs are required. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

2057 KiB  
Review
Sympathetic Overactivity in Chronic Kidney Disease: Consequences and Mechanisms
by Jasdeep Kaur, Benjamin E. Young and Paul J. Fadel
Int. J. Mol. Sci. 2017, 18(8), 1682; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081682 - 02 Aug 2017
Cited by 92 | Viewed by 8130
Abstract
The incidence of chronic kidney disease (CKD) is increasing worldwide, with more than 26 million people suffering from CKD in the United States alone. More patients with CKD die of cardiovascular complications than progress to dialysis. Over 80% of CKD patients have hypertension, [...] Read more.
The incidence of chronic kidney disease (CKD) is increasing worldwide, with more than 26 million people suffering from CKD in the United States alone. More patients with CKD die of cardiovascular complications than progress to dialysis. Over 80% of CKD patients have hypertension, which is associated with increased risk of cardiovascular morbidity and mortality. Another common, perhaps underappreciated, feature of CKD is an overactive sympathetic nervous system. This elevation in sympathetic nerve activity (SNA) not only contributes to hypertension but also plays a detrimental role in the progression of CKD independent of any increase in blood pressure. Indeed, high SNA is associated with poor prognosis and increased cardiovascular morbidity and mortality independent of its effect on blood pressure. This brief review will discuss some of the consequences of sympathetic overactivity and highlight some of the potential pathways contributing to chronically elevated SNA in CKD. Mechanisms leading to chronic sympathoexcitation in CKD are complex, multifactorial and to date, not completely understood. Identification of the mechanisms and/or signals leading to sympathetic overactivity in CKD are crucial for development of effective therapeutic targets to reduce the increased cardiovascular risk in this patient group. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

897 KiB  
Review
Personalized Medicine: New Perspectives for the Diagnosis and the Treatment of Renal Diseases
by Anna Gluba-Brzózka, Beata Franczyk, Robert Olszewski, Maciej Banach and Jacek Rysz
Int. J. Mol. Sci. 2017, 18(6), 1248; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061248 - 10 Jun 2017
Cited by 1 | Viewed by 5175
Abstract
The prevalence of renal diseases is rising and reaching 5–15% of the adult population. Renal damage is associated with disturbances of body homeostasis and the loss of equilibrium between exogenous and endogenous elements including drugs and metabolites. Studies indicate that renal diseases are [...] Read more.
The prevalence of renal diseases is rising and reaching 5–15% of the adult population. Renal damage is associated with disturbances of body homeostasis and the loss of equilibrium between exogenous and endogenous elements including drugs and metabolites. Studies indicate that renal diseases are influenced not only by environmental but also by genetic factors. In some cases the disease is caused by mutation in a single gene and at that time severity depends on the presence of one or two mutated alleles. In other cases, renal disease is associated with the presence of alteration within a gene or genes, but environmental factors are also necessary for the development of disease. Therefore, it seems that the analysis of genetic aspects should be a natural component of clinical and experimental studies. The goal of personalized medicine is to determine the right drug, for the right patient, at the right time. Whole-genome examinations may help to change the approach to the disease and the patient resulting in the creation of “personalized medicine” with new diagnostic and treatment strategies designed on the basis of genetic background of each individual. The identification of high-risk patients in pharmacogenomics analyses will help to avoid many unwarranted side effects while optimizing treatment efficacy for individual patients. Personalized therapies for kidney diseases are still at the preliminary stage mainly due to high costs of such analyses and the complex nature of human genome. This review will focus on several areas of interest: renal disease pathogenesis, diagnosis, treatment, rate of progression and the prediction of prognosis. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

721 KiB  
Review
Cholesterol Crystal Embolism and Chronic Kidney Disease
by Xuezhu Li, George Bayliss and Shougang Zhuang
Int. J. Mol. Sci. 2017, 18(6), 1120; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061120 - 24 May 2017
Cited by 35 | Viewed by 12178
Abstract
Renal disease caused by cholesterol crystal embolism (CCE) occurs when cholesterol crystals become lodged in small renal arteries after small pieces of atheromatous plaques break off from the aorta or renal arteries and shower the downstream vascular bed. CCE is a multisystemic disease [...] Read more.
Renal disease caused by cholesterol crystal embolism (CCE) occurs when cholesterol crystals become lodged in small renal arteries after small pieces of atheromatous plaques break off from the aorta or renal arteries and shower the downstream vascular bed. CCE is a multisystemic disease but kidneys are particularly vulnerable to atheroembolic disease, which can cause an acute, subacute, or chronic decline in renal function. This life-threatening disease may be underdiagnosed and overlooked as a cause of chronic kidney disease (CKD) among patients with advanced atherosclerosis. CCE can result from vascular surgery, angiography, or administration of anticoagulants. Atheroembolic renal disease has various clinical features that resemble those found in other kidney disorders and systemic diseases. It is commonly misdiagnosed in clinic, but confirmed by characteristic renal biopsy findings. Therapeutic options are limited, and prognosis is considered to be poor. Expanding knowledge of atheroembolic renal disease due to CCE opens perspectives for recognition, diagnosis, and treatment of this cause of progressive renal insufficiency. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

4802 KiB  
Review
Chronic Kidney Disease and Exposure to Nephrotoxic Metals
by Sarah E. Orr and Christy C. Bridges
Int. J. Mol. Sci. 2017, 18(5), 1039; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18051039 - 12 May 2017
Cited by 255 | Viewed by 12405
Abstract
Chronic kidney disease (CKD) is a common progressive disease that is typically characterized by the permanent loss of functional nephrons. As injured nephrons become sclerotic and die, the remaining healthy nephrons undergo numerous structural, molecular, and functional changes in an attempt to compensate [...] Read more.
Chronic kidney disease (CKD) is a common progressive disease that is typically characterized by the permanent loss of functional nephrons. As injured nephrons become sclerotic and die, the remaining healthy nephrons undergo numerous structural, molecular, and functional changes in an attempt to compensate for the loss of diseased nephrons. These compensatory changes enable the kidney to maintain fluid and solute homeostasis until approximately 75% of nephrons are lost. As CKD continues to progress, glomerular filtration rate decreases, and remaining nephrons are unable to effectively eliminate metabolic wastes and environmental toxicants from the body. This inability may enhance mortality and/or morbidity of an individual. Environmental toxicants of particular concern are arsenic, cadmium, lead, and mercury. Since these metals are present throughout the environment and exposure to one or more of these metals is unavoidable, it is important that the way in which these metals are handled by target organs in normal and disease states is understood completely. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

424 KiB  
Review
Hypoxia, HIF, and Associated Signaling Networks in Chronic Kidney Disease
by Jing Liu, Qingqing Wei, Chunyuan Guo, Guie Dong, Yu Liu, Chengyuan Tang and Zheng Dong
Int. J. Mol. Sci. 2017, 18(5), 950; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18050950 - 30 Apr 2017
Cited by 82 | Viewed by 5467
Abstract
The pathogenesis of chronic kidney disease (CKD) is complex and apparently multifactorial. Hypoxia or decrease in oxygen supply in kidney tissues has been implicated in CKD. Hypoxia inducible factors (HIF) are a small family of transcription factors that are mainly responsive to hypoxia [...] Read more.
The pathogenesis of chronic kidney disease (CKD) is complex and apparently multifactorial. Hypoxia or decrease in oxygen supply in kidney tissues has been implicated in CKD. Hypoxia inducible factors (HIF) are a small family of transcription factors that are mainly responsive to hypoxia and mediate hypoxic response. HIF plays a critical role in renal fibrosis during CKD through the modulation of gene transcription, crosstalk with multiple signaling pathways, epithelial-mesenchymal transition, and epigenetic regulation. Moreover, HIF also contributes to the development of various pathological conditions associated with CKD, such as anemia, inflammation, aberrant angiogenesis, and vascular calcification. Treatments targeting HIF and related signaling pathways for CKD therapy are being developed with promising clinical benefits, especially for anemia. This review presents an updated analysis of hypoxia response, HIF, and their associated signaling network involved in the pathogenesis of CKD. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

405 KiB  
Review
Genetics of Congenital Anomalies of the Kidney and Urinary Tract: The Current State of Play
by Valentina P. Capone, William Morello, Francesca Taroni and Giovanni Montini
Int. J. Mol. Sci. 2017, 18(4), 796; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18040796 - 11 Apr 2017
Cited by 85 | Viewed by 8866
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are the most frequent form of malformation at birth and represent the cause of 40–50% of pediatric and 7% of adult end-stage renal disease worldwide. The pathogenesis of CAKUT is based on the disturbance [...] Read more.
Congenital anomalies of the kidney and urinary tract (CAKUT) are the most frequent form of malformation at birth and represent the cause of 40–50% of pediatric and 7% of adult end-stage renal disease worldwide. The pathogenesis of CAKUT is based on the disturbance of normal nephrogenesis, secondary to environmental and genetic causes. Often CAKUT is the first clinical manifestation of a complex systemic disease, so an early molecular diagnosis can help the physician identify other subtle clinical manifestations, significantly affecting the management and prognosis of patients. The number of sporadic CAKUT cases explained by highly penetrant mutations in a single gene may have been overestimated over the years and a genetic diagnosis is missed in most cases, hence the importance of identifying new genetic approaches which can help unraveling the vast majority of unexplained CAKUT cases. The aim of our review is to clarify the current state of play and the future perspectives of the genetic bases of CAKUT. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

1009 KiB  
Review
Emerging Roles of IL-33/ST2 Axis in Renal Diseases
by Wei-Yu Chen, Lung-Chih Li and Jenq-Lin Yang
Int. J. Mol. Sci. 2017, 18(4), 783; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18040783 - 07 Apr 2017
Cited by 43 | Viewed by 6897
Abstract
Renal diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), have a great impact on health care systems worldwide. Similar to cardiovascular diseases, renal diseases are inflammatory diseases involving a variety of cytokines. Primary causes of renal injury include ischemia, uremic [...] Read more.
Renal diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), have a great impact on health care systems worldwide. Similar to cardiovascular diseases, renal diseases are inflammatory diseases involving a variety of cytokines. Primary causes of renal injury include ischemia, uremic toxins, bacteremia, or nephrotoxicity. Inflammation represents an important component following kidney injury. Interleukin (IL)-33 is a member of the IL-1 cytokine family, which is widely expressed in epithelial barrier tissues and endothelial cells, and mediates both tissue inflammation and repair responses. IL-33 is released as a nuclear alarmin in response to tissue damage and triggers innate and adaptive immune responses by binding to its receptor, suppression of tumorigenicity 2 (ST2). Recent evidence from clinical and experimental animal studies indicates that the IL-33/ST2 axis is involved in the pathogenesis of CKD, renal graft injury, systemic lupus nephritis, and AKI. In this review, we discuss the pathological and tissue reparative roles of the IL-33/ST2 pathway in different types of renal diseases. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Graphical abstract

485 KiB  
Review
Developmental Origins of Chronic Kidney Disease: Should We Focus on Early Life?
by You-Lin Tain and Chien-Ning Hsu
Int. J. Mol. Sci. 2017, 18(2), 381; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18020381 - 11 Feb 2017
Cited by 71 | Viewed by 8580
Abstract
Chronic kidney disease (CKD) is becoming a global burden, despite recent advances in management. CKD can begin in early life by so-called “developmental programming” or “developmental origins of health and disease” (DOHaD). Early-life insults cause structural and functional changes in the developing kidney, [...] Read more.
Chronic kidney disease (CKD) is becoming a global burden, despite recent advances in management. CKD can begin in early life by so-called “developmental programming” or “developmental origins of health and disease” (DOHaD). Early-life insults cause structural and functional changes in the developing kidney, which is called renal programming. Epidemiological and experimental evidence supports the proposition that early-life adverse events lead to renal programming and make subjects vulnerable to developing CKD and its comorbidities in later life. In addition to low nephron endowment, several mechanisms have been proposed for renal programming. The DOHaD concept opens a new window to offset the programming process in early life to prevent the development of adult kidney disease, namely reprogramming. Here, we review the key themes on the developmental origins of CKD. We have particularly focused on the following areas: evidence from human studies support fetal programming of kidney disease; insight from animal models of renal programming; hypothetical mechanisms of renal programming; alterations of renal transcriptome in response to early-life insults; and the application of reprogramming interventions to prevent the programming of kidney disease. Full article
(This article belongs to the Special Issue Advances in Chronic Kidney Disease 2017)
Show Figures

Figure 1

Back to TopTop