Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23.337
Filter
1.
Sci Rep ; 14(1): 18146, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103417

ABSTRACT

Urate nephropathy, a common complication of hyperuricemia, has garnered increasing attention worldwide. However, the exact pathogenesis of this condition remains unclear. Currently, inflammation is widely accepted as the key factor in urate nephropathy. Therefore, the aim of this study was to elucidate the interaction of lincRNA-p21/AIF-1/CMPK2/NLRP3 via exosomes in urate nephropathy. This study evaluated the effect of lincRNA-p21/AIF-1/CMPK2/NLRP3 using clinical data collected from patients with urate nephropathy and human renal tubular epithelial cells (HK2) cultured with different concentrations of urate. In clinical research section, the level of lincRNA-p21/AIF-1 in exosomes of urine in patients with hyperuricemia or urate nephropathy was found to be increased, particularly in patients with urate nephropathy. In vitro study section, the level of exosomes, inflammation, autophagy, and apoptosis was increased in HK2 cells induced by urate. Additionally, the expression of lincRNA-p21, AIF-1, CMPK2, and NLRP3 was upregulated in exosomes and HK2 cells. Furthermore, manipulating the activity of lincRNA-p21, AIF-1, CMPK2, and NLRP3 through overexpression or interference vectors regulated the level of inflammation, autophagy, and apoptosis in HK2 cells. In conclusion, the pathway of lincRNA-p21/AIF-1/CMPK2/NLRP3 contributed to inflammation, autophagy, and apoptosis of human renal tubular epithelial cell induced by urate via exosomes. Additionally, the specific exosomes in urine might serve as novel biomarkers for urate nephropathy.


Subject(s)
Apoptosis , Autophagy , Epithelial Cells , Exosomes , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Long Noncoding , Uric Acid , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uric Acid/metabolism , Exosomes/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Signal Transduction , Inflammation/metabolism , Inflammation/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Cell Line , Male , Apoptosis Inducing Factor/metabolism , Female , Middle Aged , Hyperuricemia/metabolism , Hyperuricemia/urine , Calcium-Binding Proteins , Microfilament Proteins
2.
PLoS One ; 19(7): e0306479, 2024.
Article in English | MEDLINE | ID: mdl-38959226

ABSTRACT

The histomorphological features of normal kidneys in cats and dogs have been revealed despite the high susceptibility of cats to tubulointerstitial damage. Herein, the histological characteristics of the two species were compared. Cytoplasmic lipid droplets (LDs) were abundant in the proximal convoluted tubules (PCTs) of cats aged 23-27 months but scarce in dogs aged 24-27 months. LDs were rarely observed in the distal tubules (DTs) and collecting ducts (CDs) of either species, as visualized by the expression of Tamm-Horsfall protein 1, calbindin-D28K, and aquaporin 2. The occupational area ratio of proximal tubules (PTs) in the renal cortex was higher, but that of DTs or CDs was significantly lower in adult cats than in dogs. Single PT epithelial cells were larger, but PCT, DT, and CD lumens were significantly narrower in adult cats than in dogs. Unlike adults, young cats at 6 months exhibited significantly abundant cytoplasmic LDs in proximal straight tubules, indicating lipid metabolism-related development. Histochemistry of the 21 lectins also revealed variations in glycosylation across different renal tubules and CDs in both species. Sodium-glucose cotransporter 2 was expressed only in PTs, excluding the proximal straight tubules with few LDs in adult cats or the PCTs of young cats and adult dogs. These findings are crucial for understanding species-specific characteristics of renal histomorphology and pathogenesis.


Subject(s)
Kidney Tubules, Collecting , Species Specificity , Animals , Dogs , Cats , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Female , Lipid Droplets/metabolism
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 488-493, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38952087

ABSTRACT

Objective To identify immune-related transcription factors (TFs) in renal glomeruli and tubules from diabetic kidney disease (DKD) patients by bioinformatics analysis. Methods Gene expression datasets from GEO (GSE30528, GSE30529) and RNA sequencing (RNA-seq) data from the Karolinska Kidney Research Center were used. Gene set enrichment analysis (GSEA) was conducted to examine differences in immune-related gene expression in the glomeruli and tubules (DKD) patients. To identify immune-related genes (IRGs) and TFs, differential expression analysis was carried out using the Limma and DESeq2 software packages. Key immune-related TFs were pinpointed through co-expression analysis. The interaction network between TFs and IRGs was constructed using the STRING database and Cytoscape software. Furthermore, the Nephroseq database was employed to investigate the correlation between the identified TFs and clinical-pathological features. Results When compared to normal control tissues, significant differences in the expression of immune genes were observed in both the glomeruli and tubules of individuals with Diabetic Kidney Disease (DKD). Through differential and co-expression analysis, 50 immune genes and 9 immune-related transcription factors (TFs) were identified in the glomeruli. In contrast, 131 immune response genes (IRGs) and 41 immune-related TFs were discovered in the renal tubules. The protein-protein interaction (PPI) network highlighted four key immune-related TFs for the glomeruli: Interferon regulatory factor 8 (IRF8), lactotransferrin (LTF), CCAAT/enhancer binding protein alpha (CEBPA), and Runt-related transcription factor 3 (RUNX3). For the renal tubules, the key immune-related TFs were FBJ murine osteosarcoma viral oncogene homolog B (FOSB), nuclear receptor subfamily 4 group A member 1 (NR4A1), IRF8, and signal transducer and activator of transcription 1 (STAT1). These identified TFs demonstrated a significant correlation with the glomerular filtration rate (GFR), highlighting their potential importance in the pathology of DKD. Conclusion Bioinformatics analysis identifies potential genes associated with DKD pathogenesis and immune dysregulation. Further validation of the expression and function of these genes may contribute to immune-based therapeutic research for DKD.


Subject(s)
Computational Biology , Diabetic Nephropathies , Transcription Factors , Humans , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Diabetic Nephropathies/metabolism , Transcription Factors/genetics , Computational Biology/methods , Gene Expression Profiling , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Gene Regulatory Networks , Kidney Tubules/immunology , Kidney Tubules/metabolism
4.
Mol Biol Rep ; 51(1): 790, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990390

ABSTRACT

INTRODUCTION: Creating induced pluripotent stem cells (iPSCs) from somatic cells of patients with genetic diseases offers a pathway to generate disease-specific iPSCs carrying genetic markers. Differentiating these iPSCs into renal tubular cells can aid in understanding the pathophysiology of rare inherited renal tubular diseases through cellular experiments. MATERIALS AND METHODS: Two Japanese patients with Pseudohypoparathyroidism (PHP), a 49-year-old woman and a 71-year-old man, were studied. iPSC-derived tubular cells were established from their peripheral blood mononuclear cells (PBMCs). We examined changes in intracellular and extracellular cyclic adenosine monophosphate (cAMP) levels in these cells in response to parathyroid hormone (PTH) stimulation. RESULTS: Renal tubular cells, differentiated from iPSCs of a healthy control (648A1), showed a PTH-dependent increase in both intracellular and extracellular cAMP levels. However, the renal tubular cells derived from the PHP patients' iPSCs showed inconsistent changes in cAMP levels upon PTH exposure. CONCLUSION: We successfully created disease-specific iPSCs from PHP patients' PBMCs, differentiated them into tubular cells, and replicated the distinctive response of the disease to PTH in vitro. This approach could enhance our understanding of the pathophysiology of inherited renal tubular diseases and contribute to developing effective treatments.


Subject(s)
Cell Differentiation , Cyclic AMP , Induced Pluripotent Stem Cells , Kidney Tubules , Leukocytes, Mononuclear , Parathyroid Hormone , Pseudohypoparathyroidism , Humans , Parathyroid Hormone/pharmacology , Parathyroid Hormone/metabolism , Induced Pluripotent Stem Cells/metabolism , Pseudohypoparathyroidism/genetics , Pseudohypoparathyroidism/metabolism , Female , Cell Differentiation/drug effects , Male , Cyclic AMP/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Middle Aged , Aged , Leukocytes, Mononuclear/metabolism , Cells, Cultured
5.
Cell Death Dis ; 15(7): 473, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956064

ABSTRACT

Damage to renal tubular epithelial cells (RTECs) signaled the onset and progression of sepsis-associated acute kidney injury (SA-AKI). Recent research on mitochondria has revealed that mitophagy plays a crucial physiological role in alleviating injury to RTECs and it is suppressed progressively by the inflammation response in SA-AKI. However, the mechanism by which inflammation influences mitophagy remains poorly understood. We examined how macrophage migration inhibitory factor (MIF), a pro-inflammatory protein, influences the PINK1-Parkin pathway of mitophagy by studying protein-protein interactions when MIF was inhibited or overexpressed. Surprisingly, elevated levels of MIF were found to directly bind to PINK1, disrupting its interaction with Parkin. This interference hindered the recruitment of Parkin to mitochondria and impeded the initiation of mitophagy. Furthermore, this outcome led to significant apoptosis of RTECs, which could, however, be reversed by an MIF inhibitor ISO-1 and/or a new mitophagy activator T0467. These findings highlight the detrimental impact of MIF on renal damage through its disruption of the interaction between PINK1 and Parkin, and the therapeutic potential of ISO-1 and T0467 in mitigating SA-AKI. This study offers a fresh perspective on treating SA-AKI by targeting MIF and mitophagy.


Subject(s)
Acute Kidney Injury , Macrophage Migration-Inhibitory Factors , Mitophagy , Protein Kinases , Sepsis , Ubiquitin-Protein Ligases , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Kinases/metabolism , Sepsis/complications , Sepsis/metabolism , Animals , Humans , Mitochondria/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Apoptosis , Protein Binding , Male , Intramolecular Oxidoreductases/metabolism
6.
Kidney Int ; 106(2): 185-188, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39032963

ABSTRACT

Acute kidney injury is still associated with high morbidity and mortality. Reichardt et al. investigated DNA-binding protein-A (Ybx3) in acute kidney injury induced by ischemia-reperfusion injury and found that mice lacking Ybx3 have altered mitochondrial function and increased antioxidant activity, making them more resistant to ischemia-reperfusion injury-acute kidney injury. The study highlights a new role of the multifaceted protein DNA-binding protein-A, which could be potentially therapeutically exploited.


Subject(s)
Acute Kidney Injury , Epithelial Cells , Kidney Tubules , Reperfusion Injury , Animals , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/etiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mice , Kidney Tubules/metabolism , Kidney Tubules/pathology , Kidney Tubules/cytology , Humans , Mitochondria/metabolism , Oxidative Stress
7.
Ren Fail ; 46(2): 2376929, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39022902

ABSTRACT

The transient receptor potential canonical 6 (TRPC6) channel, a nonselective cation channel that allows the passage of Ca2+, plays an important role in renal diseases. TRPC6 is activated by Ca2+ influx, oxidative stress, and mechanical stress. Studies have shown that in addition to glomerular diseases, TRPC6 can contribute to renal tubular disorders, such as acute kidney injury, renal interstitial fibrosis, and renal cell carcinoma (RCC). However, the tubule-specific physiological functions of TRPC6 have not yet been elucidated. Its pathophysiological role in ischemia/reperfusion (I/R) injury is debatable. Thus, TRPC6 may have dual roles in I/R injury. TRPC6 induces renal fibrosis and immune cell infiltration in a unilateral ureteral obstruction (UUO) mouse model. Additionally, TRPC6 overexpression may modify G2 phase transition, thus altering the DNA damage checkpoint, which can cause genomic instability and RCC tumorigenesis and can control the proliferation of RCC cells. This review highlights the importance of TRPC6 in various conditions of the renal tubular system. To better understand certain renal disorders and ultimately identify new therapeutic targets to improve patient care, the pathophysiology of TRPC6 must be clarified.


Subject(s)
TRPC6 Cation Channel , Humans , TRPC6 Cation Channel/metabolism , TRPC6 Cation Channel/genetics , Animals , Kidney Tubules/pathology , Kidney Tubules/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Reperfusion Injury/metabolism , Fibrosis , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Mice , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Oxidative Stress , Kidney Diseases/metabolism , Kidney Diseases/etiology
8.
Clin Sci (Lond) ; 138(15): 963-973, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39076039

ABSTRACT

Renal tubules have potential to regenerate and repair after mild-to-moderate injury. Proliferation of tubular epithelial cells represents the initial step of this reparative process. Although for many years, it was believed that proliferating cells originated from a pre-existing intra-tubular stem cell population, there is now consensus that surviving tubular epithelial cells acquire progenitor properties to regenerate the damaged kidney. Scattered tubular-like cells (STCs) are dedifferentiated adult renal tubular epithelial cells that arise upon injury and contribute to renal self-healing and recovery by replacing lost neighboring tubular epithelial cells. These cells are characterized by the co-expression of the stem cell surface markers CD133 and CD24, as well as mesenchymal and kidney injury markers. Previous studies have shown that exogenous delivery of STCs ameliorates renal injury and dysfunction in murine models of acute kidney injury, underscoring the regenerative potential of this endogenous repair system. Although STCs contain fewer mitochondria than their surrounding terminally differentiated tubular epithelial cells, these organelles modulate several important cellular functions, and their integrity and function are critical to preserve the reparative capacity of STCs. Recent data suggest that the microenviroment induced by cardiovascular risk factors, such as obesity, hypertension, and renal ischemia may compromise STC mitochondrial integrity and function, limiting the capacity of these cells to repair injured renal tubules. This review summarizes current knowledge of the contribution of STCs to kidney repair and discusses recent insight into the key role of mitochondria in modulating STC function and their vulnerability in the setting of cardiovascular disease.


Subject(s)
Mitochondria , Regeneration , Humans , Mitochondria/metabolism , Animals , Regeneration/physiology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Acute Kidney Injury/pathology , Kidney/metabolism , Epithelial Cells/metabolism
9.
Chem Biol Interact ; 399: 111152, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39025289

ABSTRACT

Cadmium (Cd), a prevalent environmental contaminant, has attracted widespread attention due to its serious health hazards. Ferroptosis is a form of iron-dependent oxidative cell death that contributes to the development of various kidney diseases. However, the mechanisms underlying the occurrence of ferroptosis in Cd-induced renal tubular epithelial cells (TECs) have not been fully elucidated. Hereby, both in-vitro and in-vivo experiments were established to elucidate this issue. In this study, we found that Cd elicited accumulation of lipid peroxides due to intracellular ferrous ion (Fe2+) overload and glutathione depletion, contributing to ferroptosis. Inhibition of ferroptosis via chelation of Fe2+ or reduction of lipid peroxidation can significantly mitigate Cd-induced cytotoxicity. Renal transcriptome analysis revealed that the activation of heme oxygenase 1 (HO-1) was closely related to ferroptosis in Cd-induced TECs injury. Cd-induced ferroptosis and resultant TECs injury are significantly alleviated due to HO-1 inhibition, demonstrating the crucial role of HO-1 in Cd-triggered ferroptosis. Further studies showed that accumulation of lipid peroxides due to iron overload and mitochondrial ROS (mtROS) generation was responsible for HO-1-triggered ferroptosis in Cd-induced cytotoxicity. In conclusion, the current study demonstrates that excessively upregulating HO-1 promotes iron overload and mtROS overproduction to trigger ferroptosis in Cd-induced TECs injury, highlighting that targeting HO-1-mediated ferroptosis may provide new ideas for preventing Cd-induced nephrotoxicity.


Subject(s)
Cadmium , Epithelial Cells , Ferroptosis , Heme Oxygenase-1 , Iron , Kidney Tubules , Mitochondria , Reactive Oxygen Species , Ferroptosis/drug effects , Cadmium/toxicity , Heme Oxygenase-1/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Kidney Tubules/cytology , Kidney Tubules/pathology , Iron/metabolism , Mice , Lipid Peroxidation/drug effects , Cell Line , Male , Humans , Glutathione/metabolism , Mice, Inbred C57BL
10.
Mol Med ; 30(1): 112, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085830

ABSTRACT

BACKGROUND: Patients with type 2 diabetes often face early tubular injury, necessitating effective treatment strategies. This study aimed to evaluate the impact of the SGLT2 inhibitor empagliflozin on early tubular injury biomarkers in type 2 diabetes patients with normoalbuminuria. METHODS: A randomized controlled clinical study comprising 54 patients selected based on specific criteria was conducted. Patients were divided into an intervention group (empagliflozin, n = 27) and a control group (n = 27) and treated for 6 weeks. Tubular injury biomarkers KIM-1 and NGAL were assessed pre- and post-treatment. RESULTS: Both groups demonstrated comparable baseline characteristics. Post-treatment, fasting and postprandial blood glucose levels decreased similarly in both groups. The intervention group exhibited better improvements in total cholesterol, low-density lipoprotein, and blood uric acid levels. Renal function indicators, including UACR and eGFR, showed greater enhancements in the intervention group. Significant reductions in KIM-1 and NGAL were observed in the intervention group. CONCLUSION: Treatment with empagliflozin in type 2 diabetes patients with normoalbuminuria led to a notable decrease in tubular injury biomarkers KIM-1 and NGAL. These findings highlight the potential of SGLT2 inhibitors in early tubular protection, offering a new therapeutic approach.


Subject(s)
Benzhydryl Compounds , Biomarkers , Diabetes Mellitus, Type 2 , Glucosides , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Male , Female , Middle Aged , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Hepatitis A Virus Cellular Receptor 1/metabolism , Blood Glucose , Aged , Lipocalin-2/blood , Adult , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Diabetic Nephropathies/prevention & control
11.
Cell Commun Signal ; 22(1): 357, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987851

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is highly prevalent worldwide, and its global burden is substantial and growing. CKD displays a number of features of accelerated senescence. Tubular cell senescence is a common biological process that contributes to CKD progression. Tubulointerstitial inflammation is a driver of tubular cell senescence and a common characteristic of CKD. However, the mechanism by which the interstitial inflammation drives tubular cell senescence remains unclear. This paper aims to explore the role of exosomal miRNAs derived from macrophages in the development of tubular cell senescence. METHODS: Among the identified inflammation-related miRNAs, miR-155 is considered to be one of the most important miRNAs involved in the inflammatory response. Macrophages, the primary immune cells that mediate inflammatory processes, contain a high abundance of miR-155 in their released exosomes. We assessed the potential role of miR-155 in tubular cell senescence and renal fibrosis. We subjected miR-155-/- mice and wild-type controls, as well as tubular epithelial cells (TECs), to angiotensin II (AngII)-induced kidney injury. We assessed kidney function and injury using standard techniques. TECs were evaluated for cell senescence and telomere dysfunction in vivo and in vitro. Telomeres were measured by the fluorescence in situ hybridization. RESULTS: Compared with normal controls, miR-155 was up-regulated in proximal renal tubule cells in CKD patients and mouse models of CKD. Moreover, the expression of miR-155 was positively correlated with the extent of renal fibrosis, eGFR decline and p16INK4A expression. The overexpression of miR-155 exacerbated tubular senescence, evidenced by increased detection of p16INK4A/p21expression and senescence-associated ß-galactosidase activity. Notably, miR-155 knockout attenuates renal fibrosis and tubule cell senescence in vivo. Interestingly, once released, macrophages-derived exosomal miR-155 was internalized by TECs, leading to telomere shortening and dysfunction through targeting TRF1. A dual-luciferase reporter assay confirmed that TRF1 was the direct target of miR-155. Thus, our study clearly demonstrates that exosomal miR-155 may mediate communication between macrophages and TECs, subsequently inducing telomere dysfunction and senescence in TECs. CONCLUSIONS: Our work suggests a new mechanism by which macrophage exosomes are involved in the development of tubule senescence and renal fibrosis, in part by delivering miR-155 to target TRF1 to promote telomere dysfunction. Our study may provide novel strategies for the treatment of AngII-induced kidney injury.


Subject(s)
Cellular Senescence , Epithelial Cells , Exosomes , Kidney Tubules , Macrophages , MicroRNAs , Telomere , MicroRNAs/genetics , MicroRNAs/metabolism , Cellular Senescence/genetics , Exosomes/metabolism , Exosomes/genetics , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Macrophages/metabolism , Kidney Tubules/pathology , Kidney Tubules/metabolism , Mice , Telomere/genetics , Telomere/metabolism , Humans , Mice, Inbred C57BL , Male , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Fibrosis/genetics , Angiotensin II
12.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1135-1140, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977343

ABSTRACT

OBJECTIVE: To investigate the protective effect of dexmedetomidine (DEX) against erastin-induced ferroptosis in human renal tubular epithelial cells (HK-2 cells) and explore the underlying mechanism. METHODS: HK-2 cells were treated with erastin alone or in combination with different concentrations (2.5, 5.0 and 10 µmol/L) of DEX, and the changes in cell viability were observed using CCK-8 assay. To explore the mechanism by which DEX inhibits erastin-induced ferroptosis, HK-2 cells were treated with erastin, erastin+10 µmol/L DEX, or erastin+10 µmol/L DEX+ML385 (a Nrf2 inhibitor), after which the cell viability was assessed. The level of intracellular Fe2+ was detected by cell ferrous iron colorimetric assay kit, and flow cytometry was performed to detect reactive oxygen species (ROS); MDA and reduced glutathione assay kits were used to detect the contents of MDA and GSH in the cells; The expressions of Nrf2, HO-1 and GPX4 proteins were detected by Western blotting. RESULTS: Erastin treatment significantly inhibited the viability of the cells, decreased GSH content, and increased intracellular levels of Fe2+, ROS and MDA. The combined treatment with 10 µmol/L DEX markedly increased the viability of the cells, increased GSH content, reduced the levels of Fe2+, ROS and MDA, and upregulated the protein expressions of Nrf2, HO-1 and GPX4 in the cells. The application of ML385 obviously blocked the protective effect of DEX and caused significant inhibition of the Nrf2/HO-1/GPX4 pathway, decreased the cell viability and GSH content, and increased the levels of Fe2+, ROS and MDA in HK-2 cells. CONCLUSION: The protective effect of DEX against erastin-induced ferroptosis of HK-2 cells is probably mediated by activation of the Nrf2/HO-1/GPX4 pathway to inhibit oxidative stress.


Subject(s)
Cell Survival , Dexmedetomidine , Epithelial Cells , Ferroptosis , Heme Oxygenase-1 , Kidney Tubules , NF-E2-Related Factor 2 , Phospholipid Hydroperoxide Glutathione Peroxidase , Reactive Oxygen Species , Humans , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Dexmedetomidine/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Kidney Tubules/cytology , Kidney Tubules/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Cell Line , Cell Survival/drug effects , Heme Oxygenase-1/metabolism , Signal Transduction/drug effects , Piperazines/pharmacology
13.
Nat Commun ; 15(1): 5731, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977708

ABSTRACT

Neuropilin-1 (NRP1), a co-receptor for various cytokines, including TGF-ß, has been identified as a potential therapeutic target for fibrosis. However, its role and mechanism in renal fibrosis remains elusive. Here, we show that NRP1 is upregulated in distal tubular (DT) cells of patients with transplant renal insufficiency and mice with renal ischemia-reperfusion (I-R) injury. Knockout of Nrp1 reduces multiple endpoints of renal injury and fibrosis. We find that Nrp1 facilitates the binding of TNF-α to its receptor in DT cells after renal injury. This signaling results in a downregulation of lysine crotonylation of the metabolic enzyme Cox4i1, decreases cellular energetics and exacerbation of renal injury. Furthermore, by single-cell RNA-sequencing we find that Nrp1-positive DT cells secrete collagen and communicate with myofibroblasts, exacerbating acute kidney injury (AKI)-induced renal fibrosis by activating Smad3. Dual genetic deletion of Nrp1 and Tgfbr1 in DT cells better improves renal injury and fibrosis than either single knockout. Together, these results reveal that targeting of NRP1 represents a promising strategy for the treatment of AKI and subsequent chronic kidney disease.


Subject(s)
Acute Kidney Injury , Fibrosis , Mice, Knockout , Neuropilin-1 , Receptor, Transforming Growth Factor-beta Type I , Reperfusion Injury , Smad3 Protein , Neuropilin-1/metabolism , Neuropilin-1/genetics , Animals , Humans , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Male , Tumor Necrosis Factor-alpha/metabolism , Signal Transduction , Mice, Inbred C57BL , Kidney Tubules/pathology , Kidney Tubules/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Collagen/metabolism
14.
Sci Rep ; 14(1): 15635, 2024 07 07.
Article in English | MEDLINE | ID: mdl-38972889

ABSTRACT

This study aimed to elucidate the influence of miR-483-3p on human renal tubular epithelial cells (HK-2) under high glucose conditions and to understand its mechanism. Human proximal tubular epithelial cells (HK-2) were exposed to 50 mmol/L glucose for 48 h to establish a renal tubular epithelial cell injury model, denoted as the high glucose group (HG group). Cells were also cultured for 48 h in a medium containing 5.5 mmol/L glucose, serving as the low glucose group. Transfection was performed in various groups: HK-2 + low glucose (control group), high glucose (50 mM) (HG group), high glucose + miR-483-3p mimics (HG + mimics group), high glucose +miR-483-3p inhibitor (HG + inhibitor group), and corresponding negative controls. Real-time quantitative polymerase chain reaction (qPCR) assessed the mRNA expression of miR-483-3p, bax, bcl-2, and caspase-3. Western blot determined the corresponding protein levels. Proliferation was assessed using the CCK-8 assay, and cell apoptosis was analyzed using the fluorescence TUNEL method. Western blot and Masson's staining were conducted to observe alterations in cell fibrosis post miR-483-3p transfection. Furthermore, a dual-luciferase assay investigated the targeting relationship between miR-483-3p and IGF-1. The CCK8 assay demonstrated that the HG + mimics group inhibited HK-2 cell proliferation, while the fluorescent TUNEL method revealed induced cell apoptosis in this group. Conversely, the HG + inhibitor group promoted cell proliferation and suppressed cell apoptosis. The HG + mimics group upregulated mRNA and protein expression of pro-apoptotic markers (bax and caspase-3), while downregulating anti-apoptotic marker (bcl-2) expression. In contrast, the HG + inhibitor group showed opposite effects. Collagen I and FN protein levels were significantly elevated in the HG + mimics group compared to controls (P < 0.05). Conversely, in the HG + inhibitor group, the protein expression of Collagen I and FN was notably reduced compared to the HG group (P < 0.05). The dual luciferase reporter assay confirmed that miR-483-3p could inhibit the luciferase activity of IGF-1's 3'-UTR region (P < 0.05). miR-483-3p exerts targeted regulation on IGF-1, promoting apoptosis and fibrosis in renal tubular epithelial cells induced by high glucose conditions.


Subject(s)
Apoptosis , Cell Proliferation , Epithelial Cells , Glucose , Insulin-Like Growth Factor I , Kidney Tubules , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Glucose/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Insulin-Like Growth Factor I/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Line , Kidney Tubules/metabolism , Kidney Tubules/cytology , Gene Expression Regulation/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Caspase 3/metabolism , Caspase 3/genetics
15.
J Biochem Mol Toxicol ; 38(8): e23768, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39015062

ABSTRACT

Nephrotoxicity remains a major adverse reaction of the anticancer drug cisplatin (CDDP) chemotherapy, which is an important risk factor for chronic renal disease. Ginsenoside Rh2 from Panax ginseng has been shown to protect against CDDP-induced nephrotoxicity in vivo, but its pharmacological effect on renal tubular epithelial cells is not clearly understood. This study examined the molecular mechanisms underlying the nephroprotective effects of Rh2 on CDDP-induced HK-2 cells and acute kidney injury (AKI) mice. As a result of Rh2 treatment, CDDP-induced HK-2 cells showed increased cell viability and reduced lactate dehydrogenase release. Moreover, Rh2 ameliorated CDDP-induced mitochondrial membrane potential, increased antioxidant enzyme activities, and reduced pro-inflammatory cytokine expression to reduce damage. Rh2 inhibited apoptosis and enhanced the antioxidant capacity of HK-2 cells by reducing proteins associated with endoplasmic reticulum (ER) stress, as well as by attenuating tunicamycin-induced ER stress. In addition, treatment of CDDP-induced AKI mice with Rh2 substantially reduced blood urea nitrogen and serum creatinine levels, attenuated histological damage of kidney. Further, Rh2 also improved kidney function by inhibiting ER stress to support in vitro findings. These results consistently demonstrated that Rh2 protects renal tubular epithelial cells from CDDP-induced nephrotoxicity and apoptosis by restoring ER homeostasis, which might suggest a therapeutic potential and providing new insights into AKI alternative therapies.


Subject(s)
Acute Kidney Injury , Cisplatin , Endoplasmic Reticulum Stress , Epithelial Cells , Ginsenosides , Kidney Tubules , Ginsenosides/pharmacology , Cisplatin/adverse effects , Cisplatin/toxicity , Endoplasmic Reticulum Stress/drug effects , Animals , Mice , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/metabolism , Humans , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/drug therapy , Male , Cell Line , Apoptosis/drug effects , Mice, Inbred C57BL
16.
Ren Fail ; 46(2): 2361089, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38874156

ABSTRACT

As a pattern recognition receptor, Toll-like receptor 4 (TLR4) is crucial for the development and progression of acute kidney injury (AKI). This study aims to explore whether the deubiquitinase Usp9x influences the TLR4/NF-B pathway to cause sepsis-induced acute kidney injury (S-AKI). The model of AKI was established in Sprague-Dawley rats using the cecal ligation and puncture (CLP) method, while renal tubular epithelial cell NRK-52E was stimulated with lipopolysaccharide (LPS) in vitro. All plasmids were transfected into NRK-52E cells according to the indicated group. The deubiquitinase of TLR4 was predicted by the online prediction software Ubibrowser. Subsequently, Western blot and Pearson correlation analysis identified Usp9x protein as a potential candidate. Co-IP analysis verified the interaction between TLR4 and Usp9x. Further research revealed that overexpression of Usp9x inhibited degradation of TLR4 protein by downregulating its ubiquitination modification levels. Both in vivo and in vitro experiments observed that interference with Usp9x effectively alleviated the inflammatory response and apoptosis of renal tubular epithelial cells (RTECs) induced by CLP or LPS, whereas overexpression of TLR4 reversed this situation. Transfection with sh-Usp9x in NRK-52E cells suppressed the expression of proteins associated with the TLR4/NF-κB pathway induced by LPS. Moreover, the overexpression of TLR4 reversed the effect of sh-Usp9x transfection. Therefore, the deubiquitinase Usp9x interacts with TLR4, leading to the upregulation of its expression through deubiquitination modification, and the activation of the TLR4/NF-κB signaling pathway, thereby promoting inflammation and apoptosis in renal tubular epithelial cells and contributing to sepsis-induced acute kidney injury.


Subject(s)
Acute Kidney Injury , Apoptosis , Epithelial Cells , Inflammation , Kidney Tubules , NF-kappa B , Rats, Sprague-Dawley , Sepsis , Signal Transduction , Toll-Like Receptor 4 , Ubiquitin Thiolesterase , Animals , Toll-Like Receptor 4/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Sepsis/complications , Sepsis/metabolism , NF-kappa B/metabolism , Rats , Epithelial Cells/metabolism , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/cytology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Male , Inflammation/metabolism , Disease Models, Animal , Cell Line , Lipopolysaccharides , Ubiquitination
17.
Int J Biol Sci ; 20(8): 3061-3075, 2024.
Article in English | MEDLINE | ID: mdl-38904010

ABSTRACT

Renal fibrosis is the common pathway in the progression of chronic kidney disease (CKD). Acyloxyacyl hydrolase (AOAH) is expressed in various phagocytes and is highly expressed in proximal tubular epithelial cells (PTECs). Research shows that AOAH plays a critical role in infections and chronic inflammatory diseases, although its role in kidney injury is unknown. Here, we found that AOAH deletion led to exacerbated kidney injury and fibrosis after folic acid (FA) administration, which was reversed by overexpression of Aoah in kidneys. ScRNA-seq revealed that Aoah-/- mice exhibited increased subpopulation of CD74+ PTECs, though the percentage of total PTECs were decreased compared to WT mice after FA treatment. Additionally, exacerbated kidney injury and fibrosis seen in Aoah-/- mice was attenuated via administration of methyl ester of (S, R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (ISO-1), an inhibitor of macrophage inhibition factor (MIF) and CD74 binding. Finally, AOAH expression was found positively correlated with estimated glomerular filtration rate while negatively correlated with the degree of renal fibrosis in kidneys of CKD patients. Thus, our work indicates that AOAH protects against kidney injury and fibrosis by inhibiting renal tubular epithelial cells CD74 signaling pathways. Targeting kidney AOAH represents a promising strategy to prevent renal fibrosis progression.


Subject(s)
Carboxylic Ester Hydrolases , Macrophages , Animals , Mice , Macrophages/metabolism , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/genetics , Humans , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Renal Insufficiency, Chronic/metabolism , Mice, Inbred C57BL , Male , Histocompatibility Antigens Class II/metabolism , Folic Acid/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Fibrosis/metabolism , Mice, Knockout , Epithelial Cells/metabolism
18.
Curr Aging Sci ; 17(2): 118-126, 2024.
Article in English | MEDLINE | ID: mdl-38904154

ABSTRACT

Containing information molecules from their parent cells and inclining to fuse with targeted cells, bone marrow mesenchymal stromal cells-derived extracellular vesicles (MSCs- EV) are valuable in nanomedicine. BACKGROUND: The effects of aging on the paracrine mechanism and in the production and action of MSCs-EV and their cargos of miR-26a and siRNA-26a for the treatment of tubular renal cells under nephrotoxicity injury remain unelucidated. OBJECTIVE: The purpose of this study was to evaluate MSCs-EV of different ages and their ability to deliver the cargos of miR-26a and siRNA-26a to target renal tubular cells affected by nephrotoxicity injury. METHODS: In a model of gentamicin-induced nephrotoxicity, renal tubular cells treated with MSCs-EV expressing or not expressing microRNA-26a were analyzed. Western blotting was utilized to evaluate cell cycle markers, and MTT assay was utilized to evaluate auto-renovation capacity. RESULTS: Tubular cells under nephrotoxicity injury showed decreased proliferative capacity, but the treatment in the tubular renal cells under nephrotoxicity injury with MSCs-EV expressing microRNA-26a showed nephroprotective effects, regardless of EV age. While the treatment with EV-mediated siRNA-26a failed to preserve the nephroprotective effects equally, regardless of age. CONCLUSION: Mesenchymal stromal cell nanovesicles carry microRNA with nephroprotective proprieties regardless of aging.


Subject(s)
Cell Proliferation , Kidney Tubules , Mesenchymal Stem Cells , MicroRNAs , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Kidney Tubules/pathology , Kidney Tubules/metabolism , Aging/metabolism , Aging/pathology , Aging/genetics , Gentamicins/toxicity , Gentamicins/adverse effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Age Factors , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Cell Line , Cells, Cultured , Paracrine Communication , Disease Models, Animal , Humans
19.
Int J Rheum Dis ; 27(6): e15210, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38837302

ABSTRACT

INTRODUCTION: The relevance of tubulo-interstitial involvement for kidney prognosis has recently been emphasized, but validated biomarkers for predicting histology are still lacking. The aim of our study was to evaluate different serum and urinary markers of tubular damage in patients with lupus nephritis (LN) and to correlate them with kidney histopathology. METHODS: A single-center retrospective study was conducted from January 2016 to December 2021. Serum and urine samples were collected on the same day of kidney biopsy and correlated with histologic data from a cohort of 15 LN patients. We analyzed the following urinary markers, adjusted for urine creatinine: beta 2-microglobulin, alpha 1-microglobulin, NGAL, uKIM-1, MCP-1, uDKK-3, and uUMOD. The serum markers sKIM-1 and sUMOD were also analyzed. RESULTS: A positive and strong correlation was observed between the degree of interstitial fibrosis (rho = 0.785, p = .001) and tubular atrophy (rho = 0.781, p = .001) and the levels of uDKK3. uUMOD also showed an inverse and moderate correlation with interstitial fibrosis (rho = -0.562, p = .037) and tubular atrophy (rho = -0.694, p = .006). Patients with >10% cortical interstitial inflammation had higher levels of uKIM-1 [4.9 (3.9, 5.5) vs. 0.8 (0.6, 1.5) mcg/mg, p = .001], MCP-1 [3.8 (2. 3, 4.2) vs. 0.7 (0.3, 1.2) mcg/mg, p = .001], sKIM-1 [9.2 (5.9, 32.7) vs. 1.4 (0, 3.5) pg/mL, p = .001], and lower sUMOD [8.7 (0, 39.7) vs. 46.1 (35.7, 53) ng/mL, p = .028]. CONCLUSION: The use of specific urinary and serum biomarkers of tubular dysfunction or injury may help to predict certain histologic parameters in LN patients.


Subject(s)
Biomarkers , Kidney Tubules , Lupus Nephritis , Humans , Lupus Nephritis/urine , Lupus Nephritis/blood , Lupus Nephritis/pathology , Lupus Nephritis/diagnosis , Biomarkers/blood , Biomarkers/urine , Female , Male , Retrospective Studies , Adult , Kidney Tubules/pathology , Biopsy , Predictive Value of Tests , Middle Aged , Fibrosis , Atrophy , Young Adult
20.
BMC Nephrol ; 25(1): 192, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849771

ABSTRACT

OBJECTIVE: Contrast media (CM) is a commonly applied drug in medical examination and surgery. However, contrast-induced acute kidney injury (CIAKI) poses a severe threat to human life and health. Notably, the CUT-like homeobox 1 (CUX1) gene shows protective effects in a variety of cells. Therefore, the objective of this study was to provide a new target for the treatment of CIAKI through exploring the role and possible molecular mechanism of CUX1 in CIAKI. METHOD: Blood samples were collected from 20 patients with CIAKI and healthy volunteers. Human kidney 2 (HK-2) cells were incubated with 200 mg/mL iohexol for 6 h to establish a contrast-induced injury model of HK-2 cells. Subsequently, qRT-PCR was used to detect the relative mRNA expression of CUX1; CCK-8 and flow cytometry to assess the proliferation and apoptosis of HK-2 cells; the levels of IL(interleukin)-1ß, tumor necrosis factor alpha (TNF-α) and malondialdehyde (MDA) in cells and lactate dehydrogenase (LDH) activity in cell culture supernatant were detect; and western blot to observe the expression levels of CUX1 and the PI3K/AKT signaling pathway related proteins [phosphorylated phosphoinositide 3-kinase (p-PI3K), PI3K, phosphorylated Akt (p-AKT), AKT]. RESULTS: CUX1 expression was significantly downregulated in blood samples of patients with CIAKI and contrast-induced HK-2 cells. Contrast media (CM; iohexol) treatment significantly reduced the proliferation of HK-2 cells, promoted apoptosis, stimulated inflammation and oxidative stress that caused cell damage. CUX1 overexpression alleviated cell damage by significantly improving the proliferation level of HK-2 cells induced by CM, inhibiting cell apoptosis, and reducing the level of LDH in culture supernatant and the expression of IL-1ß, TNF-α and MDA in cells. CM treatment significantly inhibited the activity of PI3K/AKT signaling pathway activity. Nevertheless, up-regulating CUX1 could activate the PI3K/AKT signaling pathway activity in HK-2 cells induced by CM. CONCLUSION: CUX1 promotes cell proliferation, inhibits apoptosis, and reduces inflammation and oxidative stress in CM-induced HK-2 cells to alleviate CM-induced damage. The mechanism of CUX1 may be correlated with activation of the PI3K/AKT signaling pathway.


Subject(s)
Acute Kidney Injury , Apoptosis , Contrast Media , Epithelial Cells , Homeodomain Proteins , Kidney Tubules , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Humans , Apoptosis/drug effects , Signal Transduction/drug effects , Contrast Media/adverse effects , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Kidney Tubules/pathology , Kidney Tubules/metabolism , Cell Line , Transcription Factors/metabolism , Male , Iohexol , Female , Cell Proliferation/drug effects , Middle Aged , Repressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL