Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.598
Filter
1.
Ren Fail ; 46(2): 2357246, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38832490

ABSTRACT

OBJECTIVE: A two-sample Mendelian randomization (MR) analysis was performed to elucidate the causal impact of celiac disease on the risk of chronic kidney disease (CKD). METHODS: The study comprised data from three genome-wide association studies involving individuals of European ancestry. The study groups included participants with celiac disease (n = 24,269), CKD (n = 117,165), and estimated glomerular filtration rate levels based on serum creatinine (eGFRcrea, n = 133,413). We employed four widely recognized causal inference algorithms: MR-Egger, inverse variance weighted (IVW), weighted median, and weighted mode. To address potential issues related to pleiotropy and overall effects, MR-Egger regression and the MR-PRESSO global test were performed. Heterogeneity was assessed using Cochran's Q test. RESULTS: We identified 14 genetic variants with genome-wide significance. The MR analysis provided consistent evidence across the various methodologies, supporting a causal relationship between celiac disease and an elevated risk of CKD (odds ratio (OR)IVW = 1.027, p = 0.025; ORweighted median = 1.028, P = 0.049; ORweighted mode = 1.030, p = 0.044). Furthermore, we observed a causal link between celiac disease and a decreased eGFRcrea (ORIVW = 0.997, P = 2.94E-06; ORweighted median = 0.996, P = 1.68E-05; ORweighted mode = 0.996, P = 3.11E-04; ORMR Egger = 0.996, P = 5.00E-03). We found no significant evidence of horizontal pleiotropy, heterogeneity, or bias based on MR-Egger regression, MR-PRESSO, and Cochran's Q test. CONCLUSION: The results of this study indicate a causal relationship between celiac disease and an increased risk of CKD.


Subject(s)
Celiac Disease , Genome-Wide Association Study , Glomerular Filtration Rate , Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Celiac Disease/genetics , Celiac Disease/complications , Renal Insufficiency, Chronic/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Female , Male , Risk Factors
2.
Clin Exp Med ; 24(1): 94, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38703294

ABSTRACT

Prior research has established associations between immune cells, inflammatory proteins, and chronic kidney disease (CKD). Our Mendelian randomization study aims to elucidate the genetic causal relationships among these factors and CKD. We applied Mendelian randomization using genetic variants associated with CKD from a large genome-wide association study (GWAS) and inflammatory markers from a comprehensive GWAS summary. The causal links between exposures (immune cell subtypes and inflammatory proteins) and CKD were primarily analyzed using the inverse variance-weighted, supplemented by sensitivity analyses, including MR-Egger, weighted median, weighted mode, and MR-PRESSO. Our analysis identified both absolute and relative counts of CD28 + CD45RA + CD8 + T cell (OR = 1.01; 95% CI = 1.01-1.02; p < 0.001, FDR = 0.018) (OR = 1.01; 95% CI = 1.00-1.01; p < 0.001, FDR = 0.002), CD28 on CD39 + CD8 + T cell(OR = 0.97; 95% CI = 0.96-0.99; p < 0.001, FDR = 0.006), CD16 on CD14-CD16 + monocyte (OR = 1.02; 95% CI = 1.01-1.03; p < 0.001, FDR = 0.004) and cytokines, such as IL-17A(OR = 1.11, 95% CI = 1.06-1.16, p < 0.001, FDR = 0.001), and LIF-R(OR = 1.06, 95% CI = 1.02-1.10, p = 0.005, FDR = 0.043) that are genetically predisposed to influence the risk of CKD. Moreover, the study discovered that CKD itself may causatively lead to alterations in certain proteins, including CST5(OR = 1.16, 95% CI = 1.09-1.24, p < 0.001, FDR = 0.001). No evidence of reverse causality was found for any single biomarker and CKD. This comprehensive MR investigation supports a genetic causal nexus between certain immune cell subtypes, inflammatory proteins, and CKD. These findings enhance the understanding of CKD's immunological underpinnings and open avenues for targeted treatments.


Subject(s)
Genome-Wide Association Study , Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/immunology , Inflammation Mediators/metabolism , Genetic Predisposition to Disease
3.
Cell Commun Signal ; 22(1): 264, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734696

ABSTRACT

BACKGROUND: Traditional biomarkers of chronic kidney disease (CKD) detect the disease in its late stages and hardly predict associated vascular damage. Integrin-linked kinase (ILK) is a scaffolding protein and a serine/threonine protein kinase that plays multiple roles in several pathophysiological processes during renal damage. However, the involvement of ILK as a biomarker of CKD and its associated vascular problems remains to be fully elucidated. METHODS: CKD was induced by an adenine-rich diet for 6 weeks in mice. We used an inducible ILK knockdown mice (cKD-ILK) model to decrease ILK expression. ILK content in mice's peripheral blood mononuclear cells (PBMCs) was determined and correlated with renal function parameters and with the expression of ILK and fibrosis and inflammation markers in renal and aortic tissues. Also, the expression of five miRNAs that target ILK was analyzed in whole blood of mice. RESULTS: The adenine diet increased ILK expression in PBMCs, renal cortex, and aortas, and creatinine and urea nitrogen concentrations in the plasma of WT mice, while these increases were not observed in cKD-ILK mice. Furthermore, ILK content in PBMCs directly correlated with renal function parameters and with the expression of renal and vascular ILK and fibrosis and inflammation markers. Finally, the expression of the five miRNAs increased in the whole blood of adenine-fed mice, although only four correlated with plasma urea nitrogen, and of those, three were downregulated in cKD-ILK mice. CONCLUSIONS: ILK, in circulating mononuclear cells, could be a potential biomarker of CKD and CKD-associated renal and vascular damage.


Subject(s)
Biomarkers , Kidney , Leukocytes, Mononuclear , Protein Serine-Threonine Kinases , RNA, Messenger , Renal Insufficiency, Chronic , Animals , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Leukocytes, Mononuclear/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Biomarkers/metabolism , Biomarkers/blood , Mice , Kidney/pathology , Kidney/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/blood , MicroRNAs/metabolism , Disease Models, Animal , Fibrosis
4.
PLoS One ; 19(5): e0302526, 2024.
Article in English | MEDLINE | ID: mdl-38739575

ABSTRACT

Chronic Kidney Disease patients under hemodialysis have high morbidity rate, which tends to considerably affect their health-related quality of life. Multiple studies that have made use of different questionnaries report the poor life quality of this patient group. The research in hand implemented the Mind Genomics Approach as a method to asses the health-related quality of life of hemodialysis patients, while relying on conjoint measurements to group individuals with similar patterns of responses to a certain mindset. The study is conducted in 3 clinics with 219 patients. It uncovers three clusters or mindsets: Mindset 1- Feels guardedly optimistic but worried about money, Mindset 2-Feels strongly positive because the state guarantees and the family supports, Mindset 3-Feels positive only about money. Based on the analysis of the collected data, the findings of this study suggest that the quality of life in hemodialysis patients is highly correlated to their financial status. The current study is one of the few first attempts to apply Mind Genomics in medical settings and the first, to our knowledge, in hemodialysis centers. This technology might enable healthcare proffesionals to provide personalized psychological treatment and additional social support to patients, which in turn could improve their clinical outcomes. The study is an example of using technology as a service.


Subject(s)
Genomics , Quality of Life , Renal Dialysis , Humans , Renal Dialysis/psychology , Male , Female , Middle Aged , Genomics/methods , Adult , Aged , Surveys and Questionnaires , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/psychology , Renal Insufficiency, Chronic/genetics
5.
Kidney Int ; 105(6): 1162-1164, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777400

ABSTRACT

In this commentary, a novel approach to the reclassification of chronic kidney disease is reviewed. In the revisited study, the investigators identify 4 distinct subtypes of kidney disease derived from an unbiased self-organizing map of transcriptomic data from kidney biopsy samples. These molecular subtypes then are characterized by biologic cell processes, clinical and histopathologic features, urinary proteomics, and disease progression. The strengths and limitations of the self-organizing map approach are assessed; the prognostic, diagnostic, and therapeutic implications are considered briefly.


Subject(s)
Disease Progression , Kidney , Proteomics , Renal Insufficiency, Chronic , Transcriptome , Humans , Prognosis , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/urine , Proteomics/methods , Kidney/pathology , Biopsy , Gene Expression Profiling , Biomarkers/analysis , Biomarkers/urine
6.
PLoS One ; 19(5): e0302853, 2024.
Article in English | MEDLINE | ID: mdl-38768139

ABSTRACT

BACKGROUND: Chronic Kidney Disease (CKD) and Metabolic dysfunction-associated steatohepatitis (MASH) are metabolic fibroinflammatory diseases. Combining single-cell (scRNAseq) and spatial transcriptomics (ST) could give unprecedented molecular disease understanding at single-cell resolution. A more comprehensive analysis of the cell-specific ligand-receptor (L-R) interactions could provide pivotal information about signaling pathways in CKD and MASH. To achieve this, we created an integrative analysis framework in CKD and MASH from two available human cohorts. RESULTS: The analytical framework identified L-R pairs involved in cellular crosstalk in CKD and MASH. Interactions between cell types identified using scRNAseq data were validated by checking the spatial co-presence using the ST data and the co-expression of the communicating targets. Multiple L-R protein pairs identified are known key players in CKD and MASH, while others are novel potential targets previously observed only in animal models. CONCLUSION: Our study highlights the importance of integrating different modalities of transcriptomic data for a better understanding of the molecular mechanisms. The combination of single-cell resolution from scRNAseq data, combined with tissue slide investigations and visualization of cell-cell interactions obtained through ST, paves the way for the identification of future potential therapeutic targets and developing effective therapies.


Subject(s)
Renal Insufficiency, Chronic , Single-Cell Analysis , Transcriptome , Humans , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Ligands , Gene Expression Profiling , Cell Communication/genetics , Fatty Liver/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Signal Transduction
7.
Sci Rep ; 14(1): 11530, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773208

ABSTRACT

Recent reports have shown the feasibility of measuring biological age from DNA methylation levels in blood cells from specific regions identified by machine learning, collectively known as the epigenetic clock or DNA methylation clock. While extensive research has explored the association of the DNA methylation clock with cardiovascular diseases, cancer, and Alzheimer's disease, its relationship with kidney diseases remains largely unexplored. In particular, it is unclear whether the DNA methylation clock could serve as a predictor of worsening kidney function. In this pilot study involving 20 subjects, we investigated the association between the DNA methylation clock and subsequent deterioration of renal function. Additionally, we noninvasively evaluated DNA damage in urinary shedding cells using a previously reported method to examine the correlation with the DNA methylation clock and worsening kidney function. Our findings revealed that patients with an accelerated DNA methylation clock exhibited increased DNA damage in urinary shedding cells, along with a higher rate of eGFR decline. Moreover, in cases of advanced CKD (G4-5), the DNA damage in urinary shedding cells was significantly increased, highlighting the interplay between elevated DNA damage and eGFR decline. This study suggests the potential role of the DNA methylation clock and urinary DNA damage as predictive markers for the progression of chronic kidney disease.


Subject(s)
DNA Damage , DNA Methylation , Glomerular Filtration Rate , Renal Insufficiency, Chronic , Humans , Pilot Projects , Male , Female , Middle Aged , Aged , Renal Insufficiency, Chronic/urine , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Disease Progression , Biomarkers/urine , Kidney/metabolism , Kidney/pathology , Epigenesis, Genetic
8.
J Pharm Biomed Anal ; 245: 116197, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38723558

ABSTRACT

The dysregulated levels of branched chain amino acids (BCAA) contribute to renal fibrosis in chronic kidney disease (CKD), yet specific analysis of BCAA contents and how they are regulated still remain unclear. It is therefore of great scientific interest to understand BCAA catabolism in CKD and develop a sensitive method for simultaneous determination of individual BCAA and their metabolites branched chain α-ketoacids (BCKA). In this work, the important role of BCAA metabolism that drives renal fibrosis in the process of CKD was first revealed by using transcriptomics. The key target genes controlling BCAA metabolism were then validated, that is, mRNA levels of BCKDHA and BCKDHB, the regulating rate-limiting enzymes during BCAA metabolism were abnormally reduced by quantitative PCR (qPCR), and a similar drop-off trend of protein expression of BCKDH, HIBCH and MCCC2 that are closely related to BCAA metabolism was also confirmed by western blotting. Furthermore, we established a novel strategy that simultaneously determines 6 individual BCAA and BCKA in serum and tissue. The method based on dansylhydrazine derivatization and ultra-high performance liquid chromatography-tandem triple quadrupole mass spectrometry (UHPLC-QQQ-MS) achieved to simultaneously determine the contents of BCAA and BCKA, which is efficient and stable. Compared with normal rats, levels of BCAA including leucine, isoleucine and valine in serum and kidney of CKD rats was decreased, while BCKA including α-ketoisocaproic acid, α-ketomethylvaleric acid and α-ketoisovaleric acid was increased. Together, these findings revealed the abnormality of BCAA metabolism in driving the course of kidney fibrosis and CKD. Our current study sheds new light on changes in BCAA metabolism during CKD, and may facilitate development of drugs to treat CKD and renal fibrosis.


Subject(s)
Amino Acids, Branched-Chain , Fibrosis , Kidney , Rats, Sprague-Dawley , Renal Insufficiency, Chronic , Animals , Amino Acids, Branched-Chain/metabolism , Rats , Male , Chromatography, High Pressure Liquid/methods , Fibrosis/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Kidney/metabolism , Kidney/pathology , Keto Acids/metabolism , Transcriptome , Tandem Mass Spectrometry/methods , Gene Expression Profiling/methods
9.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732210

ABSTRACT

Investigating the role of podocytes in proteinuric disease is imperative to address the increasing global burden of chronic kidney disease (CKD). Studies strongly implicate increased levels of monocyte chemoattractant protein-1 (MCP-1/CCL2) in proteinuric CKD. Since podocytes express the receptor for MCP-1 (i.e., CCR2), we hypothesized that podocyte-specific MCP-1 production in response to stimuli could activate its receptor in an autocrine manner, leading to further podocyte injury. To test this hypothesis, we generated podocyte-specific MCP-1 knockout mice (Podo-Mcp-1fl/fl) and exposed them to proteinuric injury induced by either angiotensin II (Ang II; 1.5 mg/kg/d, osmotic minipump) or Adriamycin (Adr; 18 mg/kg, intravenous bolus). At baseline, there were no between-group differences in body weight, histology, albuminuria, and podocyte markers. After 28 days, there were no between-group differences in survival, change in body weight, albuminuria, kidney function, glomerular injury, and tubulointerstitial fibrosis. The lack of protection in the knockout mice suggests that podocyte-specific MCP-1 production is not a major contributor to either Ang II- or Adr-induced glomerular disease, implicating that another cell type is the source of pathogenic MCP-1 production in CKD.


Subject(s)
Angiotensin II , Chemokine CCL2 , Doxorubicin , Mice, Knockout , Podocytes , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Podocytes/metabolism , Podocytes/pathology , Podocytes/drug effects , Doxorubicin/adverse effects , Mice , Male , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Gene Deletion , Disease Models, Animal
10.
BMC Nephrol ; 25(1): 161, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730296

ABSTRACT

BACKGROUND: Previous observational studies have reported that systemic inflammatory regulators are related to the development of chronic kidney disease (CKD); however, whether these associations are causal remains unclear. The current study aimed to investigate the potential causal relationships between systemic inflammatory regulators and CKD and kidney function. METHOD: We performed bidirectional two-sample Mendelian randomization (MR) analyses to infer the underlying causal associations between 41 systemic inflammatory regulators and CKD and kidney function. The inverse-variance weighting (IVW) test was used as the primary analysis method. In addition, sensitivity analyses were executed via the Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) test and the weighted median test. RESULTS: The findings revealed 12 suggestive associations between 11 genetically predicted systemic inflammatory regulators and CKD or kidney function in the forward analyses, including 4 for CKD, 3 for blood urea nitrogen (BUN), 4 for eGFRcrea and 1 for eGFRcys. In the other direction, we identified 6 significant causal associations, including CKD with granulocyte-colony stimulating factor (GCSF) (IVW ß = 0.145; 95% CI, 0.042 to 0.248; P = 0.006), CKD with stem cell factor (SCF) (IVW ß = 0.228; 95% CI, 0.133 to 0.323; P = 2.40 × 10- 6), eGFRcrea with SCF (IVW ß =-2.90; 95% CI, -3.934 to -1.867; P = 3.76 × 10- 8), eGFRcys with GCSF (IVW ß =-1.382; 95% CI, -2.404 to -0.361; P = 0.008), eGFRcys with interferon gamma (IFNg) (IVW ß =-1.339; 95% CI, -2.313 to -0.366; P = 0.007) and eGFRcys with vascular endothelial growth factor (VEGF) (IVW ß =-1.709; 95% CI, -2.720 to -0.699; P = 9.13 × 10- 4). CONCLUSIONS: Our findings support causal links between systemic inflammatory regulators and CKD or kidney function both in the forward and reverse MR analyses.


Subject(s)
Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/blood , Glomerular Filtration Rate , Inflammation/genetics , Granulocyte Colony-Stimulating Factor/blood , Stem Cell Factor/genetics , Stem Cell Factor/blood , Kidney/metabolism , Kidney/physiopathology , Blood Urea Nitrogen
12.
Curr Opin Nephrol Hypertens ; 33(4): 375-382, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38701324

ABSTRACT

PURPOSE OF REVIEW: Parathyroid hormone (PTH) is the major peptide hormone regulator of blood calcium homeostasis. Abnormal PTH levels can be observed in patients with various congenital and acquired disorders, including chronic kidney disease (CKD). This review will focus on rare human diseases caused by PTH mutations that have provided insights into the regulation of PTH synthesis and secretion as well as the diagnostic utility of different PTH assays. RECENT FINDINGS: Over the past years, numerous diseases affecting calcium and phosphate homeostasis have been defined at the molecular level that are responsible for reduced or increased serum PTH levels. The underlying genetic mutations impair parathyroid gland development, involve the PTH gene itself, or alter function of the calcium-sensing receptor (CaSR) or its downstream signaling partners that contribute to regulation of PTH synthesis or secretion. Mutations in the pre sequence of the mature PTH peptide can, for instance, impair hormone synthesis or intracellular processing, while amino acid substitutions affecting the secreted PTH(1-84) impair PTH receptor (PTH1R) activation, or cause defective cleavage of the pro-sequence and thus secretion of a pro- PTH with much reduced biological activity. Mutations affecting the secreted hormone can alter detection by different PTH assays, thus requiring detailed knowledge of the utilized diagnostic test. SUMMARY: Rare diseases affecting PTH synthesis and secretion have offered helpful insights into parathyroid biology and the diagnostic utility of commonly used PTH assays, which may have implications for the interpretation of PTH measurements in more common disorders such as CKD.


Subject(s)
Mutation , Parathyroid Hormone , Humans , Parathyroid Hormone/metabolism , Parathyroid Hormone/blood , Parathyroid Hormone/genetics , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Parathyroid Glands/metabolism , Rare Diseases/diagnosis , Rare Diseases/genetics , Animals , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Calcium/metabolism , Genetic Predisposition to Disease , Predictive Value of Tests , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics
13.
Matrix Biol ; 130: 47-55, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723871

ABSTRACT

Proteinuria, the presence of high molecular weight proteins in the urine, is a primary indicator of chronic kidney disease. Proteinuria results from increased molecular permeability of the glomerular filtration barrier combined with saturation or defects in tubular protein reabsorption. Any solute that passes into the glomerular filtrate traverses the glomerular endothelium, the glomerular basement membrane, and the podocyte slit diaphragm. Damage to any layer of the filter has reciprocal effects on other layers to increase glomerular permeability. The GBM is thought to act as a compressible ultrafilter that has increased molecular selectivity with increased pressure due to compression that reduced the porosity of the GBM with increased pressure. In multiple forms of chronic kidney disease, crosslinking enzymes are upregulated and may act to increase GBM stiffness. Here we show that enzymatically crosslinking porcine GBM with transglutaminase increases the stiffness of the GBM and mitigates pressure-dependent reductions in molecular sieving coefficient. This was modeled mathematically using a modified membrane transport model accounting for GBM compression. Changes in the mechanical properties of the GBM may contribute to proteinuria through pressure-dependent effects on GBM porosity.


Subject(s)
Glomerular Basement Membrane , Proteinuria , Transglutaminases , Animals , Transglutaminases/metabolism , Transglutaminases/genetics , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/pathology , Swine , Proteinuria/metabolism , Pressure , Podocytes/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/genetics , Humans , Porosity
14.
BMC Med Genomics ; 17(1): 147, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807172

ABSTRACT

BACKGROUND: Human blood metabolites have demonstrated close associations with chronic kidney disease (CKD) in observational studies. Nonetheless, the causal relationship between metabolites and CKD is still unclear. This study aimed to assess the associations between metabolites and CKD risk. METHODS: We applied a two-sample Mendelian randomization (MR) analysis to evaluate relationships between 1400 blood metabolites and eight phenotypes (outcomes) (CKD, estimated glomerular filtration rate(eGFR), urine albumin to creatinine ratio, rapid progress to CKD, rapid decline of eGFR, membranous nephropathy, immunoglobulin A nephropathy, and diabetic nephropathy). The inverse variance weighted (IVW), MR-Egger, and weighted median were used to investigate the causal relationship. Sensitivity analyses were performed with Cochran's Q, MR-Egger intercept, MR-PRESSO Global test, and leave-one-out analysis. Bonferroni correction was used to test the strength of the causal relationship. RESULTS: Through the MR analysis of 1400 metabolites and eight clinical phenotypes, a total of 48 metabolites were found to be associated with various outcomes. Among them, N-acetylleucine (OR = 0.923, 95%CI: 0.89-0.957, PIVW = 1.450 × 10-5) has a strong causal relationship with lower risk of CKD after the Bonferroni-corrected test, whereas Glycine to alanine ratio has a strong causal relationship with higher risk of CKD (OR = 1.106, 95%CI: 1.063-1.151, PIVW = 5.850 × 10-7). No horizontal pleiotropy and heterogeneity were detected. CONCLUSION: Our study offers groundbreaking insights into the integration of metabolomics and genomics to reveal the pathogenesis of and therapeutic strategies for CKD. It underscores 48 metabolites as potential causal candidates, meriting further investigation.


Subject(s)
Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/genetics , Phenotype , Metabolome , Metabolomics , Glomerular Filtration Rate , Biomarkers/blood
15.
Int J Mol Sci ; 25(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38791453

ABSTRACT

Chronic kidney disease (CKD) is an emerging cause for morbidity and mortality worldwide. Acute kidney injury (AKI) can transition to CKD and finally to end-stage renal disease (ESRD). Targeted treatment is still unavailable. NF-κB signaling is associated with CKD and activated by B cell activating factor (BAFF) via BAFF-R binding. In turn, renal tubular epithelial cells (TECs) are critical for the progression of fibrosis and producing BAFF. Therefore, the direct involvement of the BAFF/BAFF-R system to the pathogenesis of CKD is conceivable. We performed non-accelerated nephrotoxic serum nephritis (NTN) as the CKD model in BAFF KO (B6.129S2-Tnfsf13btm1Msc/J), BAFF-R KO (B6(Cg)-Tnfrsf13ctm1Mass/J) and wildtype (C57BL/6J) mice to analyze the BAFF/BAFF-R system in anti-glomerular basement membrane (GBM) disease using high throughput RNA sequencing. We found that BAFF signaling is directly involved in the upregulation of collagen III as BAFF ko mice showed a reduced expression. However, these effects were not mediated via BAFF-R. We identified several upregulated genes that could explain the effects of BAFF in chronic kidney injury such as Txnip, Gpx3, Igfbp7, Ccn2, Kap, Umod and Ren1. Thus, we conclude that targeted treatment with anti-BAFF drugs such as belimumab may reduce chronic kidney damage. Furthermore, upregulated genes may be useful prognostic CKD biomarkers.


Subject(s)
B-Cell Activating Factor , B-Cell Activation Factor Receptor , Mice, Knockout , Animals , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Mice , B-Cell Activation Factor Receptor/metabolism , B-Cell Activation Factor Receptor/genetics , Signal Transduction , Mice, Inbred C57BL , Nephritis/metabolism , Nephritis/genetics , Nephritis/pathology , Gene Expression Profiling , Transcriptome , Disease Models, Animal , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Male
16.
Clin Sci (Lond) ; 138(10): 599-614, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38739452

ABSTRACT

AIM: Acute kidney injury (AKI) increases the risk for progressive chronic kidney disease (CKD). MicroRNA (miR)-486-5p protects against kidney ischemia-reperfusion (IR) injury in mice, although its long-term effects on the vasculature and development of CKD are unknown. We studied whether miR-486-5p would prevent the AKI to CKD transition in rat, and affect vascular function. METHODS: Adult male rats were subjected to bilateral kidney IR followed by i.v. injection of liposomal-packaged miR-486-5p (0.5 mg/kg). Kidney function and histologic injury were assessed after 24 h and 10 weeks. Kidney endothelial protein levels were measured by immunoblot and immunofluorescence, and mesenteric artery reactivity was determined by wire myography. RESULTS: In rats with IR, miR-486-5p blocked kidney endothelial cell increases in intercellular adhesion molecule-1 (ICAM-1), reduced neutrophil infiltration and histologic injury, and normalized plasma creatinine (P<0.001). However, miR-486-5p attenuated IR-induced kidney endothelial nitric oxide synthase (eNOS) expression (P<0.05). At 10 weeks, kidneys from rats with IR alone had decreased peritubular capillary density and increased interstitial collagen deposition (P<0.0001), and mesenteric arteries showed impaired endothelium-dependent vasorelaxation (P<0.001). These changes were inhibited by miR-486-5p. Delayed miR-486-5p administration (96 h, 3 weeks after IR) had no impact on kidney fibrosis, capillary density, or endothelial function. CONCLUSION: In rats, administration of miR-486-5p early after kidney IR prevents injury, and protects against CKD development and systemic endothelial dysfunction. These protective effects are associated with inhibition of endothelial ICAM-1 and occur despite reduction in eNOS. miR-486-5p holds promise for the prevention of ischemic AKI and its complications.


Subject(s)
Acute Kidney Injury , Intercellular Adhesion Molecule-1 , Kidney , MicroRNAs , Rats, Sprague-Dawley , Renal Insufficiency, Chronic , Reperfusion Injury , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Male , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Renal Insufficiency, Chronic/prevention & control , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Kidney/pathology , Kidney/blood supply , Kidney/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/genetics , Nitric Oxide Synthase Type III/metabolism , Rats , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism
17.
Front Endocrinol (Lausanne) ; 15: 1319753, 2024.
Article in English | MEDLINE | ID: mdl-38726345

ABSTRACT

Background: The relationship between basal metabolic rate (BMR) and Chronic kidney disease (CKD) remains unclear and controversial. In this study, we investigated the causal role of BMR in renal injury, and inversely, whether altered renal function causes changes in BMR. Methods: In this two-sample mendelian randomization (MR) study, Genetic data were accessed from published genome-wide association studies (GWAS) for BMR ((n = 454,874) and indices of renal function, i.e. estimated glomerular filtration rate (eGFR) based on creatinine (n =1, 004, 040), CKD (n=480, 698), and blood urea nitrogen (BUN) (n =852, 678) in European. The inverse variance weighted (IVW) random-effects MR method serves as the main analysis, accompanied by several sensitivity MR analyses. We also performed a reverse MR to explore the causal effects of the above indices of renal function on the BMR. Results: We found that genetically predicted BMR was negatively related to eGFR, (ß= -0.032, P = 4.95*10-12). Similar results were obtained using the MR-Egger (ß= -0.040, P = 0.002), weighted median (ß= -0.04, P= 5.35×10-11) and weighted mode method (ß= -0.05, P=9.92×10-7). Higher BMR had a causal effect on an increased risk of CKD (OR =1.36, 95% CI = 1.11-1.66, P =0.003). In reverse MR, lower eGFR was related to higher BMR (ß= -0.64, P = 2.32×10-6, IVW analysis). Bidirectional MR supports no causal association was observed between BMR and BUN. Sensitivity analyses confirmed these findings, indicating the robustness of the results. Conclusion: Genetically predicted high BMR is associated with impaired kidney function. Conversely, genetically predicted decreased eGFR is associated with higher BMR.


Subject(s)
Basal Metabolism , Genome-Wide Association Study , Glomerular Filtration Rate , Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Basal Metabolism/genetics , Kidney/metabolism , Polymorphism, Single Nucleotide , Kidney Function Tests , Male
18.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791272

ABSTRACT

Renal fibrosis (RF) stands as a pivotal pathological process in the advanced stages of chronic kidney disease (CKD), and impeding its progression is paramount for delaying the advancement of CKD. The miR-10 family, inclusive of miR-10a and miR-10b, has been implicated in the development of various fibrotic diseases. Nevertheless, the precise role of miR-10 in the development of RF remains enigmatic. In this study, we utilized both an in vivo model involving unilateral ureteral obstruction (UUO) in mice and an in vitro model employing TGF-ß1 stimulation in HK-2 cells to unravel the mechanism underlying the involvement of miR-10a/b in RF. The findings revealed heightened expression of miR-10a and miR-10b in the kidneys of UUO mice, accompanied by a substantial increase in p-Smad3 and renal fibrosis-related proteins. Conversely, the deletion of these two genes led to a notable reduction in p-Smad3 levels and the alleviation of RF in mouse kidneys. In the in vitro model of TGF-ß1-stimulated HK-2 cells, the co-overexpression of miR-10a and miR-10b fostered the phosphorylation of Smad3 and RF, while the inhibition of miR-10a and miR-10b resulted in a decrease in p-Smad3 levels and RF. Further research revealed that miR-10a and miR-10b, through binding to the 3'UTR region of Vasohibin-1 (VASH-1), suppressed the expression of VASH-1, thereby promoting the elevation of p-Smad3 and exacerbating the progression of RF. The miR-10 family may play a pivotal role in RF.


Subject(s)
Fibrosis , MicroRNAs , Signal Transduction , Smad3 Protein , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Smad3 Protein/metabolism , Smad3 Protein/genetics , Mice , Humans , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Male , Cell Line , Kidney/metabolism , Kidney/pathology , Disease Models, Animal , Kidney Diseases/metabolism , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice, Inbred C57BL , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology
19.
Zhonghua Yi Xue Za Zhi ; 104(16): 1363-1366, 2024 Apr 23.
Article in Chinese | MEDLINE | ID: mdl-38644285

ABSTRACT

Hereditary kidney disease is an important cause of chronic kidney disease in children. With the progress of genome sequencing, single-cell technology, and organoid cultures, the research on hereditary kidney disease has entered a new era. How to integrate big data resources, discover new disease-causing genes, and develop effective treatment methods will be the focus of future research. This article discusses the classification, research progress, challenges and prospects of pediatric hereditary kidney disease, so as to provide valuable insights into the research of hereditary kidney disease in children.


Subject(s)
Kidney Diseases , Humans , Child , Kidney Diseases/genetics , Renal Insufficiency, Chronic/genetics
20.
PLoS One ; 19(4): e0299605, 2024.
Article in English | MEDLINE | ID: mdl-38626061

ABSTRACT

BACKGROUND: The effect of dyslipidemia on kidney disease outcomes has been inconclusive, and it requires further clarification. Therefore, we aimed to investigate the effects of genetic factors on the association between dyslipidemia and the risk of chronic kidney disease (CKD) using polygenic risk score (PRS). METHODS: We analyzed data from 373,523 participants from the UK Biobank aged 40-69 years with no history of CKD. Baseline data included plasma levels of total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglyceride, as well as genome-wide genotype data for PRS. Our primary outcome, incident CKD, was defined as a composite of estimated glomerular filtration rate < 60 ml/min/1.73 m2 and CKD diagnosis according to International Classification of Disease-10 codes. The effects of the association between lipid levels and PRS on incident CKD were assessed using the Cox proportional hazards model. To investigate the effect of this association, we introduced multiplicative interaction terms into a multivariate analysis model and performed subgroup analysis stratified by PRS tertiles. RESULTS: In total, 4,424 participants developed CKD. In the multivariable analysis, PRS was significantly predictive of the risk of incident CKD as both a continuous variable and a categorized variable. In addition, lower total cholesterol, LDL-C, HDL-C, and higher triglyceride levels were significantly associated with the risk of incident CKD. There were interactions between triglycerides and intermediate and high PRS, and the interactions were inversely associated with the risk of incident CKD. CONCLUSIONS: This study showed that PRS presented significant predictive power for incident CKD and individuals in the low-PRS group had a higher risk of triglyceride-related incident CKD.


Subject(s)
Dyslipidemias , Renal Insufficiency, Chronic , Humans , Genetic Risk Score , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/genetics , Triglycerides , Cholesterol, HDL , Dyslipidemias/complications , Dyslipidemias/genetics , Genetic Predisposition to Disease , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...