Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 433
Filter
1.
J Biol Chem ; 299(4): 103074, 2023 04.
Article in English | MEDLINE | ID: mdl-36858200

ABSTRACT

Heparin can block pathological responses associated with diabetic nephropathy in animal models and human patients. Our previous studies showed that the interaction of heparin on the surface of rat mesangial cells (RMCs) entering G1 of cell division in hyperglycemic glucose: 1) blocked glucose uptake by glucose transporter 4; 2) inhibited cytosolic uridine diphosphate-glucose elevation that would occur within 6 h from G0/G1; and 3) prevented subsequent activation of hyaluronan synthesis in intracellular compartments and subsequent inflammatory responses. However, specific proteins that interact with heparin are unresolved. Here, we showed by live cell imaging that fluorescent heparin was rapidly internalized into the cytoplasm and then into the endoplasmic reticulum, Golgi, and nuclei compartments. Biotinylated-heparin was applied onto the surface of growth arrested G0/G1 RMCs in order to extract heparin-binding protein(s). SDS-PAGE gels showed two bands at ∼70 kDa in the extract that were absent when unlabeled heparin was used to compete. Trypsin digests of the bands were analyzed by MS and identified as calreticulin and prelamin A/C. Immunostaining with their antibodies identified the presence of calreticulin on the G0/G1 RMC cell surface. Previous studies have shown that calreticulin can be on the cell surface and can interact with the LDL receptor-related protein, which has been implicated in glucose transport by interaction with glucose transporter 4. Thus, cell surface calreticulin can act as a heparin receptor through a mechanism involving LRP1, which prevents the intracellular responses in high glucose and reprograms the cells to synthesize an extracellular hyaluronan matrix after division.


Subject(s)
Calreticulin , Cell Division , G1 Phase , Glucose , Heparin , Hyperglycemia , Mesangial Cells , Resting Phase, Cell Cycle , Animals , Humans , Rats , Calreticulin/metabolism , Cells, Cultured , Glomerular Mesangium/metabolism , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Heparin/pharmacology , Heparin/metabolism , Hyaluronic Acid/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Hyperglycemia/metabolism
2.
J Diabetes Investig ; 13(8): 1303-1315, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35482475

ABSTRACT

AIMS/INTRODUCTION: This study aimed to investigate the role and mechanism of circular ribonucleic acid nucleoporin 98 (circNUP98) in diabetic nephropathy (DN). MATERIALS AND METHODS: Human glomerular mesangial cells (HMCs) were stimulated with high glucose (HG) to imitate the growth environment of cells under the DN condition. Levels of genes and proteins were tested by quantitative reverse transcription polymerase chain reaction and western blot. Cell proliferation, apoptosis and inflammatory response were analyzed by using cell counting kit-8, flow cytometry and enzyme-linked immunosorbent assay analysis, respectively. Oxidative stress and fibrosis were evaluated by detecting the activity of reactive oxygen species, malondialdehyde, superoxide dismutase, fibronectin and collagen IV. The binding interaction between microribonucleic acid (miR)-151-3p and high mobility group AT-hook 2 (HMGA2) or circNUP98 was confirmed using dual-luciferase reporter, pull-down and ribonucleic acid immunoprecipitation assays. Exosomes were isolated by ultracentrifugation, and qualified by transmission electron microscopy, nanoparticle tracking analysis and western blot. RESULTS: CircNUP98 expression was higher in the serum of DN patients and HG-stimulated HMCs. Functionally, circNUP98 knockdown alleviated HG-induced proliferation, fibrosis, inflammatory response and oxidative stress in HMCs. Mechanistically, circNUP98 directly sponged miR-151-3p, which targeted HMGA2. Rescue experiments showed that miR-151-3p reversed the inhibitory effects of circNUP98 knockdown on HG-induced HMC dysfunction. Furthermore, miR-151-3p re-expression also led to an inhibition of the aforementioned biological behaviors, which was attenuated by HMGA2 upregulation. Besides that, CircNUP98 was found to be packaged into exosomes of DN, and exosomal circNUP98 possessed diagnostic value for DN patients. CONCLUSION: CircNUP98 knockdown alleviates HG-induced proliferation, fibrosis inflammation and oxidative stress in HMCs by regulating the miR-151-3p-HMGA2 axis, which might provide a potential approach for DN therapeutics.


Subject(s)
Diabetic Nephropathies , HMGA2 Protein , Mesangial Cells , MicroRNAs , Oxidative Stress , RNA, Circular , Cell Proliferation , Diabetic Nephropathies/metabolism , Fibrosis , Glucose/pharmacology , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , Inflammation/metabolism , Mesangial Cells/cytology , Mesangial Cells/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA, Circular/genetics
3.
Oxid Med Cell Longev ; 2022: 7502632, 2022.
Article in English | MEDLINE | ID: mdl-35126820

ABSTRACT

AIM: The study is aimed at verifying miR-154-5p and Smurf1 combination in glomerular mesangial cells regulating TGFß1/Smad3 pathway-related protein ubiquitination in the model of diabetic rats renal tissues, primary mesangial cells, and cell lines. METHODS: The diabetic SD rat model and high-glucose-cultured primary mesangial cells and cell lines were established. miR-154-5p mimic and inhibitor, Smurf1 siRNA, and TGF ß 1/Smad3 inhibitor (SB431542) were pretreated to make the TGFß1/Smad3 pathway and ubiquitin changes. Fluorescence in situ hybridization was used for the miR-154-5p renal localization; molecular biological detection was adopted for cell proliferation, renal function, urine protein, and pathway proteins. After bioinformatics predicted binding sites, luciferase and Co-IP were used to detect miRNA and protein binding. RESULTS: miR-154-5p was significantly increased and mainly concentrated in the glomerular of renal cortex in well-established diabetic rat renal tissues. Rno-miR-154-5p combined Rno-Smurf1 3' UTR, while Smurf1 combined Smad3 directly. Meanwhile, miR-154-5p regulates TGFß1/Smad3-mediated cell proliferation via Smurf1 ubiquitination. CONCLUSION: miR-154-5p regulates the TGFß1/Smads pathway through Smurf1 ubiquitination and promotes the fibrosis process of diabetic kidney disease.


Subject(s)
MicroRNAs/metabolism , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Antagomirs/metabolism , Cell Proliferation , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Fibrosis , Kidney/metabolism , Kidney/pathology , Male , Mesangial Cells/cytology , Mesangial Cells/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Smad3 Protein/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination
4.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34878095

ABSTRACT

Expansion of interstitial cells in the adult kidney is a hallmark of chronic disease, whereas their proliferation during fetal development is necessary for organ formation. An intriguing difference between adult and neonatal kidneys is that the neonatal kidney has the capacity to control interstitial cell proliferation when the target number has been reached. In this study, we define the consequences of inactivating the TGFß/Smad response in the mouse interstitial cell lineage. We find that pathway inactivation through loss of Smad4 leads to overproliferation of interstitial cells regionally in the kidney medulla. Analysis of markers for BMP and TGFß pathway activation reveals that loss of Smad4 primarily reduces TGFß signaling in the interstitium. Whereas TGFß signaling is reduced in these cells, marker analysis shows that Wnt/ß-catenin signaling is increased. Our analysis supports a model in which Wnt/ß-catenin-mediated proliferation is attenuated by TGFß/Smad to ensure that proliferation ceases when the target number of interstitial cells has been reached in the neonatal medulla.


Subject(s)
Cell Proliferation , Kidney/metabolism , Smad4 Protein/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Kidney/cytology , Kidney/growth & development , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , Smad4 Protein/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
5.
Naunyn Schmiedebergs Arch Pharmacol ; 395(1): 77-85, 2022 01.
Article in English | MEDLINE | ID: mdl-34792626

ABSTRACT

Upregulation of thrombin receptor protease-activated receptor 1 (PAR-1) is verified to contribute to chronic kidney diseases, including diabetic nephropathy; however, the mechanisms are still unclear. In this study, we investigated the effect of PAR-1 on high glucose-induced proliferation of human glomerular mesangial cells (HMCs), and explored the mechanism of PAR-1 upregulation from alteration of microRNAs. We found that high glucose stimulated proliferation of the mesangial cells whereas PAR-1 inhibition with vorapaxar attenuated the cell proliferation. Moreover, high glucose upregulated PAR-1 in mRNA level and protein expression while did not affect the enzymatic activity of thrombin in HMCs after 48 h culture. Then high glucose induced PAR-1 elevation was likely due to the alteration of the transcription or post-transcriptional processing. It was found that miR-17 family members including miR-17-5p, -20a-5p, and -93-5p were significantly decreased among the eight detected microRNAs only in high glucose-cultured HMCs, but miR-129-5p, miR-181a-5p, and miR-181b-5p were markedly downregulated in both high glucose-cultured HMCs and equivalent osmotic press control compared with normal glucose culture. So miR-20a was selected to confirm the role of miR-17 family on PAR-1 upregulation, finding that miR-20a-5p overexpression reversed the upregulation of PAR-1 in mRNA and protein levels induced by high glucose in HMCs. In summary, our finding indicated that PAR-1 upregulation mediated proliferation of glomerular mesangial cells induced by high glucose, and deficiency of miR-17 family resulted in PAR-1 upregulation.


Subject(s)
Mesangial Cells/cytology , MicroRNAs/genetics , Receptor, PAR-1/genetics , Cell Line , Cell Proliferation/drug effects , Diabetic Nephropathies/genetics , Down-Regulation , Glucose/metabolism , Humans , Lactones/pharmacology , Pyridines/pharmacology , Up-Regulation
6.
BMC Nephrol ; 22(1): 368, 2021 11 06.
Article in English | MEDLINE | ID: mdl-34742256

ABSTRACT

BACKGROUND: LncRNA NNT-AS1 (NNT-AS1) has been extensively studied as the causative agent in propagation and progression of lung and bladder cancers, and cholangiocarcinoma. However, its significance in proliferation and inflammation of diabetic nephropathy is enigmatic. This study focuses on the molecular mechanisms followed by NNT-AS1 to establish diabetic nephropathy (DN) and its potential miRNA target. METHODS: Bioinformatics analysis to identify potential miRNA target of NNT-AS1 and smad4 transcription factor was conducted using LncBase and TargetScan, and was subsequently confirmed by luciferase reporter assay. Relative quantitative expression of NNT-AS1 in human glomerular mesangial cells (HGMCs) was detected through quantitative real-time PCR and WB analysis. Cell proliferation was detected through CCK-8 assay, whereas, ELISA was conducted to evaluate the expression of inflammatory cytokines. Following this, relative expression of miR-214-5p and smad4 were confirmed through qRT-PCR and western blot analysis. RESULTS: Results from the experiments manifested up-regulated levels of NNT-AS1 and smad4 in the blood samples of DN patients as well as in HGMCs, whereas, downregulated levels of miR-214-5p were measured in the HGMCs suggesting the negative correlation between NNT-AS1 and miR-214-5p. Potential binding sites of NNT-AS1 showed miR-214-5p as its direct target and NNT-AS1 as potential absorber for this microRNA, in turn increasing the expression of transcription factor smad4. CONCLUSION: The data suggests that NNT-AS1 can be positively used as a potential biomarker and indicator of DN and causes extracellular matrix (ECM) accumulation and inflammation of human mesangial cells.


Subject(s)
Cell Proliferation , Diabetic Nephropathies/physiopathology , Extracellular Matrix/metabolism , Inflammation/physiopathology , Mesangial Cells/cytology , NADP Transhydrogenase, AB-Specific/physiology , RNA, Long Noncoding/physiology , Blood Glucose/metabolism , Diabetic Nephropathies/blood , Down-Regulation , Humans , Mesangial Cells/metabolism , MicroRNAs/blood , MicroRNAs/genetics , Mitochondrial Proteins/blood , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , NADP Transhydrogenase, AB-Specific/blood , NADP Transhydrogenase, AB-Specific/genetics , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , Smad4 Protein/blood , Smad4 Protein/genetics , Up-Regulation
7.
Anal Cell Pathol (Amst) ; 2021: 6619870, 2021.
Article in English | MEDLINE | ID: mdl-34012764

ABSTRACT

Diabetic nephropathy (DN) is an important microvascular complication of diabetes and is the main cause of end-stage renal disease. Type 2 mannose receptor C (MRC2) is a member of the mannose receptor protein family, which has been confirmed to have the ability to promote the cell migration signaling pathway and invasion. By complementary DNA chip screening and analysis, we found that the expression of MRC2 was upregulated in the kidneys of mice with diabetic nephropathy. However, the role of MRC2 in diabetic nephropathy is still unclear. This work studied the effect of MRC2 on diabetic nephropathy. After verifying the results of the chip by quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting, we used small interfering RNAs (siRNAs) to knock down the expression of MRC2 in mouse mesangial cells (MMCs) and analyzed the level of cell proliferation and apoptosis using western blotting, Cell Counting Kit-8, and flow cytometry. The results showed that the MRC2 knockdown inhibited MMC proliferation and induced cell apoptosis. These results suggest that MRC2 may be a molecular marker and a therapeutic target for diabetic nephropathy.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Diabetic Nephropathies/genetics , Disease Models, Animal , Gene Expression Regulation , Mannose-Binding Lectins/genetics , Membrane Glycoproteins/genetics , Receptors, Cell Surface/genetics , Animals , Blotting, Western , Cell Cycle/genetics , Cells, Cultured , Diabetic Nephropathies/metabolism , Female , Humans , Male , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mice , Middle Aged , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Acta Histochem ; 123(4): 151701, 2021 May.
Article in English | MEDLINE | ID: mdl-33691202

ABSTRACT

Pre-clinical animal models are needed to investigate and study kidney injuries and diseases. The rabbit kidney model is frequently used because various important parameters can be assessed with it. For example, histology and immunohistochemistry are indispensable as tissue morphology and composition can be investigated qualitatively as well as quantitatively. Here, different histological and immunohistochemical stainings were performed in the rabbit healthy naïve kidney tissue. First, overnight formalin fixation followed by paraffin embedding and cryopreservation with a subsequent 10-minute formalin fixation prior to staining were compared. Cryosections showed a more pronounced staining pattern, with clear borders at low magnifications, but blurred borders at higher magnifications. Then, antigen retrieval (AR) for paraffin embedded sections resulted in more prominent corresponding signals compared to stainings without AR. Moreover, several advantages and disadvantages of chromogenic versus immunofluorescence stainings were considered. Chromogenic staining was advantageous compared to immunofluorescence for collagen I and III, and to a minor degree for fibronectin. Finally, distinct structures, such as the pelvis, the calices, the glomeruli and tubuli, were stained in serial sections with diverse immunohistochemical stainings in order to delineate their composition. The following stainings were performed: standard Haematoxylin&Eosin and Elastica van Gieson staining, collagen I, collagen III, fibronectin, α-SMA, ki-67 and protease-activated receptor-2 (PAR-2). While chromogenic stainings of collagen I and collagen III were particularly useful to depict kidney structures in paraffin sections compared with cryosections, cryosections immunofluorescently stained for α-SMA were superior to paraffin sections, particularly at higher magnifications. With regard to specific structures, we found renal vessel walls positive for fibronectin and α-SMA, while the Bowman's capsule was only positive for fibronectin and α-SMA showed only tiny spots. The mesangial cells of the glomeruli and the distal tubuli were PAR-2 positive, while the proximal tubuli were PAR-2 negative.


Subject(s)
Immunohistochemistry , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Staining and Labeling , Animals , Female , Paraffin Embedding , Rabbits
9.
J Tradit Chin Med ; 41(1): 44-50, 2021 02.
Article in English | MEDLINE | ID: mdl-33522196

ABSTRACT

OBJECTIVE: To investigate the efficacy of Xiaokeping (XKP)-containing serum on the proliferation of high-glucose-induced mesangial cells (MCs) and the potential underlying mechanism. METHODS: XKP-containing serum was prepared by the intragastric administration of XKP in rats. HBZY-1 cells were cultured with normal glucose (NC group), high glucose (HG group), and high glucose with different XKP concentrations. Cell proliferation was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and the cell cycle distribution was detected by flow cytometry. The expression of p38 mitogen-activated protein kinase (p38MAPK) pathway components in MCs was detected by Western blotting and quantitative real-time polymerase chain reaction. RESULTS: The MC proliferation level in the high-glucose group was significantly higher than that in the normal control group, and XKP suppressed the HG-induced proliferation of MCs dose dependently. Moreover, flow cytometry revealed that XKP blocked cell cycle progression by inducing cell cycle arrest in G1 phase and inhibiting S phase entry. XKP down-regulated the protein and mRNA expression of p38MAPK in MCs (P < 0.05 vs HG). CONCLUSION: The present study demonstrated that XKP-containing serum inhibits high-glucoseinduced proliferation of MCs by causing cell cycle arrest at G1 phase and inhibiting S phase entry. The underlying mechanism involves the down-regulation of the p38MAPK signaling pathway, providing a theoretical basis for the use of XKP to treat diabetic kidney disease.


Subject(s)
Cell Proliferation/drug effects , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/physiopathology , Drugs, Chinese Herbal/administration & dosage , Glucose/adverse effects , Mesangial Cells/cytology , Mesangial Cells/drug effects , Animals , Cell Cycle/drug effects , Cell Line , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Glucose/metabolism , Humans , Male , Mesangial Cells/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Bioorg Med Chem ; 28(24): 115833, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33166928

ABSTRACT

Diabetic nephropathy (DN) is a severe microvascular complication of diabetes mellitus. High glucose has resulted in oxidative stress and following renal fibrosis as the crucial nodes of this disease. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor regulating transcription of many antioxidant genes and suppressing synthesis of extracellular matrix. To discover Nrf2 activators targeting DN, we have evaluated polypodiside using cell-based assays. The results showed polypodiside inhibited the high glucose-induced self-limited proliferation of glomerular meangial cells. Activation of Nrf2 and enhanced transcription to antioxidant response elements were observed in the presence of polypodiside. Oxidative stress and accumulation of extracellular matrix induced by high glucose in glomerular meangial cells have been ameliorated by polypodiside. Further investigations revealed the effects of polypodiside on glomerular meangial cells were associated with activation of Nrf2. Co-immunoprecipitation of Nrf2 disclosed polypodiside disrupted the Kelch-like ECH-associated protein-1 (Keap1)-Nrf2 interaction. Molecular docking elucidated polypodiside could enter the Nrf2 binding cavity of Keap1 via interacting with the residues encompassing that cavity. These findings indicate polypodiside is a Keap1-dependent Nrf2 activator affording the catabatic effects against oxidative stress and accumulation of extracellular matrix in glomerular meangial cells under high glucose.


Subject(s)
Extracellular Matrix/metabolism , Glucosides/pharmacology , NF-E2-Related Factor 2/agonists , Oxidative Stress/drug effects , Binding Sites , Cell Line , Cell Survival/drug effects , Coumaric Acids/chemistry , Drug Evaluation, Preclinical , Extracellular Matrix/drug effects , Glucose/pharmacology , Glucosides/chemistry , Glucosides/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/chemistry , Kelch-Like ECH-Associated Protein 1/metabolism , Mesangial Cells/cytology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , Polypodium/chemistry , Polypodium/metabolism , Reactive Oxygen Species/metabolism
11.
Sci Rep ; 10(1): 16005, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994511

ABSTRACT

We previously reported that the circulatory level of Asymmetric dimethylarginine (ADMA), an endogenous competitive inhibitor of nitric oxide synthase, was increased in diabetic kidney disease patients. However, the mechanism and the role of ADMA in diabetic kidney injury remain unclear. Hence, our principal aim is to investigate the causal role of ADMA in the progression of renal cell fibrosis under high glucose (HG) treatment and to delineate its signaling alterations in kidney cell injury. High Glucose/ADMA significantly increased fibrotic events including cell migration, invasion and proliferation along with fibrotic markers in the renal cells; whereas ADMA inhibition reversed the renal cell fibrosis. To delineate the central role of ADMA induced fibrotic signaling pathway and its downstream signaling, we analysed the expression levels of fibrotic markers, NOX4, ROS and ERK activity by using specific inhibitors and genetic manipulation techniques. ADMA stimulated the ROS generation along with a significant increase in NOX4 and ERK activity. Further, we observed that ADMA activated NOX-4 and ERK are involved in the extracellular matrix proteins accumulation. Also, we observed that ADMA induced ERK1/2 phosphorylation was decreased after NOX4 silencing. Our study mechanistically demonstrates that ADMA is involved in the progression of kidney cell injury under high glucose condition by targeting coordinated complex mechanisms involving the NOX4- ROS-ERK pathway.


Subject(s)
Arginine/analogs & derivatives , Diabetic Retinopathy/metabolism , Glucose/adverse effects , Kidney/pathology , NADPH Oxidase 4/metabolism , Animals , Arginine/adverse effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Kidney/cytology , Kidney/metabolism , MAP Kinase Signaling System/drug effects , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mesangial Cells/pathology , Models, Biological , Rats , Reactive Oxygen Species/metabolism
12.
Mol Med Rep ; 22(5): 3785-3794, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32901868

ABSTRACT

Circular RNAs (circRNAs) have crucial roles in various diseases; however, the mechanisms of action underlying circRNAs in the occurrence and development of diabetic nephropathy (DN) remains largely unknown. The present study investigated the differentially expressed circRNAs in the DN mice kidney cortex using circRNA sequencing and elucidated the role of circRNAs in mesangial cells. It was revealed that 40 circRNAs were unconventionally expressed, including 18 upregulated circRNAs and 22 downregulated circRNAs. Furthermore, circ_0000491 levels were significantly augmented in both DN mice and high glucose (HG, 30 mM)­induced mouse mesangial cells (MES13 cells). Knockdown of circ_0000491 significantly suppressed the increase of vimentin, fibronectin and α­smooth muscle actin, as well as collagen type I, III and IV, whilst reversing the decrease of E­cadherin in HG­induced MES13 cells. It was further revealed that circRNA_0000491 sponged miR­101b and that miR­101b directly targets TGFßRI. In addition, the expression levels of miR­101b were negatively associated with the transcriptional level of circRNA_0000491 and miR­101b inhibitors reversed the suppression of extracellular matrix (ECM)­associated protein synthesis mediated by knocking­down circRNA_0000491. In conclusion, the present study investigated the circRNA_0000491/miR­101b/TGFßRI axis in ECM accumulation and fibrosis­associated protein expression levels of mesangial cells, which suggested that circRNA_0000491 may be beneficial for the development of an effective therapeutic target for DN.


Subject(s)
Diabetic Nephropathies/genetics , Mesangial Cells/metabolism , MicroRNAs/genetics , RNA, Circular/genetics , Receptor, Transforming Growth Factor-beta Type I/drug effects , Animals , Cell Line , Diabetic Nephropathies/metabolism , Extracellular Matrix/metabolism , Glucose/adverse effects , Male , Mesangial Cells/cytology , Mesangial Cells/drug effects , Mice , Sequence Analysis, RNA , Up-Regulation
13.
J Biol Chem ; 295(42): 14262-14278, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32732288

ABSTRACT

Interaction of transforming growth factor-ß (TGFß)-induced canonical signaling with the noncanonical kinase cascades regulates glomerular hypertrophy and matrix protein deposition, which are early features of glomerulosclerosis. However, the specific target downstream of the TGFß receptor involved in the noncanonical signaling is unknown. Here, we show that TGFß increased the catalytic loop phosphorylation of platelet-derived growth factor receptor ß (PDGFRß), a receptor tyrosine kinase expressed abundantly in glomerular mesangial cells. TGFß increased phosphorylation of the PI 3-kinase-interacting Tyr-751 residue of PDGFRß, thus activating Akt. Inhibition of PDGFRß using a pharmacological inhibitor and siRNAs blocked TGFß-stimulated phosphorylation of proline-rich Akt substrate of 40 kDa (PRAS40), an intrinsic inhibitory component of mTORC1, and prevented activation of mTORC1 in the absence of any effect on Smad 2/3 phosphorylation. Expression of constitutively active myristoylated Akt reversed the siPDGFRß-mediated inhibition of mTORC1 activity; however, co-expression of the phospho-deficient mutant of PRAS40 inhibited the effect of myristoylated Akt, suggesting a definitive role of PRAS40 phosphorylation in mTORC1 activation downstream of PDGFRß in mesangial cells. Additionally, we demonstrate that PDGFRß-initiated phosphorylation of PRAS40 is required for TGFß-induced mesangial cell hypertrophy and fibronectin and collagen I (α2) production. Increased activating phosphorylation of PDGFRß is also associated with enhanced TGFß expression and mTORC1 activation in the kidney cortex and glomeruli of diabetic mice and rats, respectively. Thus, pursuing TGFß noncanonical signaling, we identified how TGFß receptor I achieves mTORC1 activation through PDGFRß-mediated Akt/PRAS40 phosphorylation to spur mesangial cell hypertrophy and matrix protein accumulation. These findings provide support for targeting PDGFRß in TGFß-driven renal fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Fibronectins/metabolism , Humans , Kidney Cortex/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
14.
Biochem Biophys Res Commun ; 529(3): 740-746, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32736701

ABSTRACT

Endocytosis by podocytes is gaining increased attention as a biologic means of removing large proteins such as serum albumin from the glomerular barrier. Some of this function has been attributed to the megalin/cubilin (Lrp2/Cubn) receptor complex and the albumin recycling protein FcRn (Fcgrt). However, whether other glomerular cells possess the potential to perform this same phenomenon or express these proteins remains uncharacterized. Mesangial cells are uniquely positioned in glomeruli and represent a cell type capable of performing several diverse functions. Here, the expression of megalin and FcRn in murine mesangial cells along with the megalin adaptor protein Dab-2 (Dab2) was shown for the first time. Cubilin mRNA expression was detected, but the absence of the cubilin partner amnionless (Amn) suggested that cubilin is minimally functional, if at all, in these cells. Mesangial cell endocytosis of albumin was characterized and shown to involve a receptor-mediated process. Albumin endocytosis was significantly impaired (p < 0.01) under inducible megalin knockdown conditions in stably transduced mesangial cells. The current work provides both the novel identification of megalin and FcRn in mesangial cells and the functional demonstration of megalin-mediated albumin endocytosis.


Subject(s)
Endocytosis , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mesangial Cells/cytology , Serum Albumin, Bovine/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cattle , Cell Line , Histocompatibility Antigens Class I/metabolism , Mesangial Cells/metabolism , Mice , Receptors, Fc/metabolism
15.
Int Immunopharmacol ; 88: 106859, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32795896

ABSTRACT

Lupus nephritis (LN) is the most serious manifestation of systemic lupus erythematosus (SLE) and a major risk of mortality. This research focused on the function of microRNA-16 (miR-16) in LN development. Fcgamma receptor II-b-deficient (Fcgr2b-/-) mice with the natural potential to develop SLE- and LN-like diseases were used. Gain- and loss-of-function studies were performed to explore the function of miR-16 in pathological symptoms in mouse kidney tissues and the proliferation of mesangial cells (SV40 MES-13). The putative downstream molecules of miR-16 were explored. Consequently, poor expression of miR-16 was found in kidney tissues. Upregulation of miR-16 inhibited tissue hyperplasia, inflammatory infiltration, glomerular injury and fibrosis but increased cell apoptosis in mouse kidney tissues, and it inhibited proliferation but promoted apoptosis of MES-13 cells as well. miR-16 directly bound to DEC2 and inactivated the TLR4 signaling. DEC2 blocked the protective roles of miR-16 in MES-13 cells. The enhanced proliferation in MES-13 cells following miR-16 inhibition was reversed by chloroquine phosphate, a TLR4 antagonist. To sum up, miR-16 was evidenced to have a potent protective capacity in LN through relieving the LN symptoms in kidney tissues and reducing proliferation of mesangial cells, during which DEC2 silencing and TLR4 signaling deficit were involved.


Subject(s)
Hyperplasia/immunology , Lupus Nephritis/immunology , Mesangial Cells/immunology , MicroRNAs/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Cytokines/metabolism , Disease Models, Animal , Fibrosis/genetics , Hyperplasia/etiology , Hyperplasia/metabolism , Inflammation/drug therapy , Inflammation/genetics , Lupus Nephritis/complications , Lupus Nephritis/pathology , Lupus Nephritis/urine , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , Protein Binding , Receptors, IgG/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism , Transcription Factors/genetics
16.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(12): 158793, 2020 12.
Article in English | MEDLINE | ID: mdl-32800850

ABSTRACT

BACKGROUND: Our previous study suggested that palmitate (PA) induces human glomerular mesangial cells (HMCs) fibrosis. However, the mechanism is not fully understood. Recent studies suggested that transient receptor potential canonical channel 6 (TRPC6)/nuclear factor of activated T cell 2 (NFAT2) played an important role in renal fibrosis. Moreover, cluster of differentiation 36 (CD36) regulated the synthesis of TPRC6 agonist diglyceride. In the present study, we investigated whether PA induced HMCs fibrosis via TRPC6/NFAT2 mediated by CD36. METHODS: A type 2 diabetic nephropathy (DN) model was established in Sprague Dawley rats, and HMCs were stimulated with PA. Lipid accumulation and free fatty acid (FFA) uptake were measured. The expression levels of TGF-ß1, p-Smad2/3, FN, TRPC6, NFAT2 and CD36 were evaluated. The intracellular calcium concentration ([Ca2+]i) was assessed. RESULTS: FFA were elevated in type 2 DN rats with kidney fibrosis in addition to NFAT2 and CD36 expression. In vitro, PA induced HMCs fibrosis, [Ca2+]i elevation and NFAT2 activation. SKF96365 or TRPC6-siRNA could attenuate PA-induced HMCs damage. By contrast, the TRPC6 activator showed the opposite effect. Moreover, NFAT2-siRNA also suppressed PA-induced HMCs fibrosis. CD36 knockdown inhibited the PA-induced [Ca2+]i elevation and NFAT2 expression. In addition, long-term treatment with PA decreased TRPC6 expression in HMCs. CONCLUSION: The results of this study demonstrated that PA could induce the activation of the [Ca2+]i/NFAT2 signaling pathway through TRPC6, which led to HMCs fibrosis. Although activation of TRPC6 attributed to CD36-mediated lipid deposition, long-term stimulation of PA may lead to negative feedback on the expression of TPRC6.


Subject(s)
CD36 Antigens/metabolism , Kidney Glomerulus/pathology , Mesangial Cells/pathology , NFATC Transcription Factors/metabolism , Palmitates/metabolism , TRPC6 Cation Channel/metabolism , Animals , Calcium/metabolism , Cell Line , Diabetic Nephropathies/metabolism , Fibrosis , Humans , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Male , Mesangial Cells/cytology , Mesangial Cells/metabolism , Rats, Sprague-Dawley
17.
Int J Mol Sci ; 21(9)2020 May 11.
Article in English | MEDLINE | ID: mdl-32403414

ABSTRACT

Combined androgen blockade using bicalutamide (Bic) is a therapeutic choice for treating prostate cancer (PCa). However, even at regular clinical dosages, Bic frequently shows adverse effects associated with cardiovascular and renal damage. Previously, we found that Bic selectively damaged mesangial cells compared to tubular cells and in an in vivo rat model, we also found renal damage caused by Bic. In the present study, a rat mesangial cell model was used to further the investigation. Results indicated that Bic enhanced lactate dehydrogenase release, reactive oxygen species (ROS) production, lysosome population and kidney injury molecule-1 and decreased N-cadherin. Bic elicited mitochondrial swelling and reduced the mitochondrial potential, resulting in severe suppression of the oxygen consumption rate (OCR), maximum respiration and ATP production. The hypoxia-inducible factor (HIF)-1 transcriptional activity and messenger RNA were significantly upregulated in dose-dependent manners. The HIF-1 protein reached a peak value at 24 h then rapidly decayed. BCL2/adenovirus E1B 19-kDa protein-interacting protein 3 and cleaved caspase-3 were dose-dependently upregulated by Bic (60 M) and that eventually led to cell apoptosis. It is suggested that Bic induces renal damage via ROS and modulates HIF-1 pathway and clinically, some protective agents like antioxidants are recommended for co-treatment.


Subject(s)
Anilides/pharmacology , Hypoxia-Inducible Factor 1/genetics , Kidney/drug effects , Mesangial Cells/drug effects , Mitochondria/drug effects , Nitriles/pharmacology , Reactive Oxygen Species/metabolism , Tosyl Compounds/pharmacology , Androgen Antagonists/pharmacology , Animals , Apoptosis/drug effects , Cadherins/metabolism , Cell Line , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1/metabolism , Kidney/metabolism , Kidney/pathology , Membrane Potential, Mitochondrial/drug effects , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mitochondria/metabolism , Rats , Up-Regulation/drug effects
18.
Clin Exp Pharmacol Physiol ; 47(7): 1203-1211, 2020 07.
Article in English | MEDLINE | ID: mdl-32077518

ABSTRACT

C1qTNF-related protein 6 (CTRP6) is a member of the CTRP family and exerts a key role in the progression of diabetes mellitus. However, the role of CTRP6 in diabetic nephropathy remains unknown. The present study was designed to examine the roles of CTRP6 in diabetic nephropathy and explore the potential molecular mechanisms. Our results showed that the expression level of CTRP6 was significantly increased in high glucose (HG)-stimulated glomerular mesangial cells (MCs). The following loss/gain-of-function assays demonstrated that CTRP6 knockdown significantly inhibited HG-induced reactive oxygen species (ROS) production in MCs. CTRP6 knockdown caused significant decreases in tumour necrosis factor-α (TNF-α), interleukin (IL)-1ß and IL-6 production levels in HG-induced MCs. Moreover, knockdown of CTRP6 inhibited HG-stimulated extracellular matrix (ECM) accumulation in MCs characterized by decreased expression and production levels of fibronectin (FN) and collagen IV (Col IV). Furthermore, CTRP6 knockdown suppressed HG-induced the activation of Akt/NF-κB pathway in MCs, while overexpression of CTRP6 exhibited the opposite effects. Treatment with LY294002, an inhibitor of Akt, reversed the induction effects of CTRP6 overexpression on ROS production, inflammation and ECM accumulation in MCs. In conclusion, these findings demonstrated that CTRP6 knockdown inhibits HG-induced ROS production, inflammation and ECM accumulation in MCs, which were mediated by the inactivation of the Akt/NF-κB pathway. The roles of CTRP6 in diabetic nephropathy provided evidence for its therapeutic potential for the treatment of diabetic nephropathy.


Subject(s)
Collagen/genetics , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Mesangial Cells/cytology , NF-kappa B/metabolism , Oxidative Stress/genetics , Proto-Oncogene Proteins c-akt/metabolism , Cell Line , Collagen/deficiency , Humans , Inflammation/genetics , Mesangial Cells/metabolism
19.
Macromol Biosci ; 20(4): e1900427, 2020 04.
Article in English | MEDLINE | ID: mdl-32077622

ABSTRACT

Multivalent nanoparticle binding to cells can be of picomolar avidity making such interactions almost as intense as those seen with antibodies. However, reducing nanoparticle design exclusively to avidity optimization by the choice of ligand and its surface density does not sufficiently account for controlling and understanding cell-particle interactions. Cell uptake, for example, is of paramount significance for a plethora of biomedical applications and does not exclusively depend on the intensity of multivalency. In this study, it is shown that the mobility of ligands tethered to particle surfaces has a substantial impact on particle fate upon binding. Nanoparticles carrying angiotensin-II tethered to highly mobile 5 kDa long poly(ethylene glycol) (PEG) chains separated by ligand-free 2 kDa short PEG chains show a superior accumulation in angiotensin-II receptor type 1 positive cells. In contrast, when ligand mobility is constrained by densely packing the nanoparticle surface with 5 kDa PEG chains only, cell uptake decreases by 50%. Remarkably, irrespective of ligand mobility and density both particle types have similar EC50 values in the 1-3 × 10-9 m range. These findings demonstrate that ligand mobility on the nanoparticle corona is an indispensable attribute to be considered in particle design to achieve optimal cell uptake via multivalent interactions.


Subject(s)
Angiotensin II/chemistry , Drug Carriers/chemistry , Mesangial Cells/drug effects , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Angiotensin II/metabolism , Animals , Biological Transport/drug effects , Cell Line , Chlorpromazine/pharmacology , Cytochalasin D/pharmacology , Gene Expression , Genistein/pharmacology , Kinetics , Ligands , Mesangial Cells/cytology , Mesangial Cells/metabolism , Nanoparticles/metabolism , Nanoparticles/ultrastructure , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Protein Binding , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism
20.
Cells ; 9(1)2020 01 17.
Article in English | MEDLINE | ID: mdl-31963523

ABSTRACT

Methylglyoxal (MGO), a highly reactive dicarbonyl compound, is a key precursor of the formation of advanced glycation end products (AGEs). MGO and MGO-AGEs were reportedly increased in patients with diabetic dysfunction, including diabetic nephropathy. The activation of glyoxalase-I (GLO-I) increases MGO and MGO-AGE detoxification. MGO-mediated glucotoxicity can also be ameliorated by MGO scavengers such as N-acetylcysteine (NAC), aminoguanidine (AG), and metformin. In this study, we noted that l-cysteine demonstrated protective effects against MGO-induced glucotoxicity in renal mesangial cells. l-cysteine prevented MGO-induced apoptosis and necrosis, together with a reduction of reactive oxygen species (ROS) production in MES13 cells. Interestingly, l-cysteine significantly reduced MGO-AGE formation and also acted as an MGO-AGE crosslink breaker. Furthermore, l-cysteine treatment accelerated MGO catabolism to D-lactate via the upregulation of GLO-I. The reduction of AGE formation and induction of AGE breakdown, following l-cysteine treatment, further supports the potential use of l-cysteine as an alternative for the therapeutic control of MGO-induced renal complications in diabetes, especially against diabetic nephropathy.


Subject(s)
Cysteine/pharmacology , Mesangial Cells/drug effects , Pyruvaldehyde/toxicity , Acetylcysteine/chemistry , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Survival/drug effects , Cysteine/chemistry , Cysteine/metabolism , Guanidines/pharmacology , Humans , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Lactoylglutathione Lyase/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mesangial Cells/cytology , Mice , Pyruvaldehyde/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL