Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Exp Clin Endocrinol Diabetes ; 130(1): 30-36, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32726814

ABSTRACT

AIMS: Our study aimed to investigate the role of long non-coding RNA ANRIL (lnc-ANRIL) knock-down in regulating cell activities, inflammation and downstream signaling pathways in mouse mesangial cellular diabetic nephropathy (DN) model.: METHODS: The mouse mesangial cells (SV40-MES13 cells) were treated with high-glucose (HG) to construct cellular DN model. Lnc-ANRIL knock-down plasmid and control knock-down plasmid were transfected into HG-treated SV40-MES13 cells as Sh-ANRIL group and Sh-NC group respectively. RESULTS: Lnc-ANRIL expression was significantly higher in HG-treated SV40-MES13 cells compared with normal glucose-treated SV40-MES13 cells and osmotic control-treated SV40-MES13 cells. Lnc-ANRIL knock-down suppressed cell proliferation and promoted cell apoptosis in HG-treated SV40-MES13 cells. As for fibrosis, lnc-ANRIL knock-down reduced fibronectin and collagen I expressions in HG-treated SV40-MES13 cells. Besides, the expressions of supernatant tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin (IL)-1ß, IL-6, IL-8 and IL-18 were reduced in Sh-ANRIL group compared with Sh-NC group. Furthermore, Wnt3, ß-catenin, p-MEK1 and p-ERK1 expressions were suppressed in Sh-ANRIL group compared with Sh-NC group, which suggested that lnc-ANRIL knock-down inhibited Wnt/ß-catenin and MEK/ERK pathways in HG-treated SV40-MES13 cells. CONCLUSIONS: Lnc-ANRIL knock-down suppresses mouse mesangial cell proliferation, fibrosis, inflammation, Wnt/ß-catenin and MEK/ERK pathways in DN.


Subject(s)
Diabetic Nephropathies/metabolism , Inflammation/metabolism , MAP Kinase Signaling System , Mesangial Cells/physiology , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway , Animals , Cell Proliferation/physiology , Fibrosis , Mice
2.
DNA Cell Biol ; 40(10): 1308-1316, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34520253

ABSTRACT

Diabetic nephropathy (DN) is a complication of diabetes, which leads to most end-stage kidney diseases and threatens health of patients. Mucin 1 (MUC1) is a heterodimeric oncoprotein, which is abnormally expressed in tumors and hematologic diseases. The aim of this study is to clarify the mechanism and role of MUC1 in DN. The mesangial cells (MCs) suffered from high glucose (HG) treatment to mimic DN in vitro. The cell proliferation was detected by Cell Counting Kit-8 assay and 5-ethynyl-2-deoxyuridine (EdU) staining assay. The expression of MUC1 and fibrosis markers: fibronectin, collagen I, and collagen IV were assessed by western blot. In this study, we demonstrated that HG treatment induced MUC1 expression in MCs. With knockdown of MUC1 or overexpressed MUC1 in MCs, the results indicated that knockdown of MUC1 inhibited MCs proliferation and reduced kidney fibrosis markers expression, including fibronectin, collagen I, and collagen IV, whereas overexpression of MUC1 led to opposite results. Mechanically, MUC1 activated signal transducers and activators of transcription (STAT) and ß-catenin signal pathway. After added AG490 (STAT inhibitor) or FH535 (ß-catenin inhibitor), blocking STAT3 and ß-catenin signal pathway attenuated MUC1-induced cell proliferation and fibronectin production in MCs. Finally, knockdown of MUC1 attenuated DN-induced kidney fibrosis in db/db mice. Therapeutic target for DN. In conclusion, MUC1 promotes MCs proliferation and kidney fibrosis in DN through activating STAT and ß-catenin signal pathway, which can help to provide a novel therapeutic target for DN.


Subject(s)
Cell Proliferation , Diabetic Nephropathies/metabolism , Mesangial Cells/metabolism , Mucin-1/metabolism , Signal Transduction , Animals , Cells, Cultured , Fibronectins/metabolism , Fibrosis , Kidney/metabolism , Kidney/pathology , Male , Mesangial Cells/physiology , Mice , Mice, Inbred C57BL , Mucin-1/genetics , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Sulfonamides/pharmacology , Tyrphostins/pharmacology , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
3.
FASEB J ; 35(7): e21751, 2021 07.
Article in English | MEDLINE | ID: mdl-34156114

ABSTRACT

Rat Thy-1 nephritis (Thy-1N) is an animal model of human mesangioproliferative glomerulonephritis (MsPGN), accompanied by glomerular mesangial cell (GMC) proliferation and extracellular matrix (ECM) deposition. Although sublytic C5b-9 formed on GMC membrane could induce cell proliferation, the mechanism is still unclear. In this study, we first demonstrated that the level of SRY related HMG-BOX gene 9 (SOX9), general control nonderepressible 5 (GCN5), fibroblast growth factor 1 (FGF1) and platelet-derived growth factor α (PDGFα) was all elevated both in the renal tissues of Thy-1N rats (in vivo) and in the GMCs (in vitro) with sublytic C5b-9 stimulation. Then, we not only discovered that sublytic C5b-9 caused GMC proliferation through increasing SOX9, GCN5, FGF1 and PDGFα expression, but also proved that SOX9 and GCN5 formed a complex and combined with FGF1 and PDGFα promoters, leading to FGF1 and PDGFα gene transcription. More importantly, GCN5 could mediate SOX9 acetylation at lysine 62 (K62) to enhance SOX9 binding to FGF1 or PDGFα promoter and promote FGF1 or PDGFα synthesis and GMC proliferation. Besides, the experiments in vivo also showed that FGF1 and PDGFα expression, GMC proliferation and urinary protein secretion in Thy-1N rats were greatly reduced by silencing renal SOX9, GCN5, FGF1 or PDGFα gene. Furthermore, the renal tissues of MsPGN patients also exhibited positive expression of these genes mentioned above. Collectively, our findings indicate that GCN5, SOX9 and FGF1/PDGFα can form an axis and play an essential role in sublytic C5b-9-triggered GMC proliferation, which might provide a novel insight into the pathogenesis of Thy-1N and MsPGN.


Subject(s)
Cell Proliferation/genetics , Cell Proliferation/physiology , Complement Membrane Attack Complex/genetics , Kidney/physiology , Mesangial Cells/physiology , Nephritis/genetics , Transcription, Genetic/genetics , Acetylation , Animals , Cell Line , Extracellular Matrix/genetics , Fibroblast Growth Factor 1/genetics , Humans , Male , Platelet-Derived Growth Factor/genetics , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , SOX9 Transcription Factor/genetics , Thy-1 Antigens/genetics , p300-CBP Transcription Factors/genetics
4.
Int Urol Nephrol ; 53(6): 1247-1254, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33942213

ABSTRACT

AIM: High glucose (HG) induces the production of transforming growth factor (TGF)-ß and reactive oxygen species, which further activates JAK/STAT signaling and promotes the synthesis of matrix proteins, contributes to the pathophysiological processes of diabetic nephropathy. This study aims to investigate the protection role of vitamin D (VD) in the kidney in high glucose condition. METHODS: Rat glomerular mesangial cells were cultured in high glucose medium, with or without VD or VD receptor (VDR) siRNAs treatment. The levels of TGF-ß and fibronectin were detected by qRT-PCR, immunoblotting and enzyme-linked immunosorbent assay (ELISA). The levels of phosphorylated JAK2, STAT1 and STAT3, and JAK/STAT signaling downstream genes were examined by immunoblotting and qRT-PCR. RESULTS: In rat glomerular mesangial cells, VD treatment can repress the tyrosine phosphorylation of JAK2, STAT1 and STAT3. VD inhibited TGF-ß and fibronectin expression which was rescued by vitamin d receptor (VDR) siRNA and STATs inhibitor perficitinib. The JAK/STAT signaling downstream protein coding genes including SOCS1, SOCS3 and type IV collagen were repressed by VD. Meanwhile, the expression of non-coding RNAs such as miR-181a, miR-181b, was repressed by VD, and the expression of miR-34a and Let-7b was upregulated by VD. CONCLUSION: Vitamin D (VD) treatment inhibits the function of HG on fibronectin production through regulating JAK/STAT pathway. These results provide direct evidences that VD protects glomerular mesangial cells from high glucose-induced injury through repressing JAK/STAT signaling, which has the potential for clinical DN treatment.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/physiology , Mesangial Cells/drug effects , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects , Vitamin D/pharmacology , Vitamins/pharmacology , Animals , Cells, Cultured , Glucose/metabolism , Male , Mesangial Cells/physiology , Rats , Rats, Sprague-Dawley
5.
Cell Immunol ; 361: 104279, 2021 03.
Article in English | MEDLINE | ID: mdl-33422698

ABSTRACT

Mesangial cells (MCs), as resident cells of the kidneys, play an important role in maintaining glomerular function. MCs are located between the capillary loops of the glomeruli and mainly support the capillary plexus, constrict blood vessels, extracellular matrix components, produce cytokines, and perform phagocytosis and clearance of macromolecular substances. When the glomerular environment changes, MCs are often affected, which can lead to functional transformation. The immune response is involved in the occurrence and development of various kidney diseases, in these diseases, antigen-presenting cells (APCs) play an important role. APCs can present antigens to T lymphocytes, causing them to become activated and proliferate. Studies have shown that MCs have phagocytic function and express APC markers on the cell surface. Additionally, MCs are stimulated by or produce various inflammatory factors to participate in the renal inflammatory response. Therefore, MCs have potential antigen presentation function and participate in the pathological changes of various kidney diseases as APCs upon activation. In this paper, by reviewing MC phagocytic function, activated MC expression of APC surface markers, and MC participation in the inflammatory response and local renal immune response, we confirm that activated MCs can act as APCs in renal disease.


Subject(s)
Mesangial Cells/immunology , Mesangial Cells/metabolism , Mesangial Cells/physiology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation , Kidney/immunology , Kidney Diseases/metabolism , Phagocytosis/immunology , T-Lymphocytes
6.
Acta Diabetol ; 58(1): 47-62, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32816106

ABSTRACT

AIMS: Glomerular mesangial cell (MC) proliferation is one of the main pathological changes in diabetic nephropathy (DN), but its mechanism needs further elaboration. The Hippo and PI3K/Akt signalling pathways are involved in the regulation of MC proliferation, but their relationship in hyperglycaemia-induced MC proliferation has not been reported. METHODS: We used db/db mice and high-glucose-cultured mesangial cells to generate a diabetic nephropathy model. An MST1-knockdown plasmid was used to identify whether the PI3K/Akt pathway is linked to the Hippo pathway through MST1. LY294002 and SC79 were used to verify the role of the PI3K/Akt signalling pathway in MC cells. RNA silencing and overexpression were performed by using YAP and PTEN-expression/knockdown plasmids to investigate the function of YAP and PTEN, respectively, in the Hippo and PI3K/Akt signalling pathways. RESULTS: By examining a potential feedback loop, we found decreased phosphorylation of MST1 and Lats1 and increased PI3K/Akt activation in db/db mice and high glucose-treated MCs, along with increased MC proliferation. The results of our gene silencing experiment proved PI3K/Akt-mediated intervention in the Hippo pathway and the regulatory effect of YAP on PI3K/Akt through PTEN. CONCLUSIONS: The Hippo pathway is inhibited under diabetic conditions, leading to YAP activation and promoting MC proliferation. The PI3K/Akt pathway is activated through the inhibitory effect of YAP on its repressor, PTEN. Finally, activation of the PI3K/Akt pathway inhibits the Hippo pathway, resulting in nuclear YAP accumulation and accelerating MC proliferation and DN formation.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Cycle Proteins/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Mesangial Cells/physiology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Dose-Response Relationship, Drug , Glucose/pharmacology , Hippo Signaling Pathway , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , YAP-Signaling Proteins
7.
Gene ; 765: 145114, 2021 Jan 10.
Article in English | MEDLINE | ID: mdl-32891769

ABSTRACT

The current study aimed to investigate the role and underlying mechanisms of circ_LARP4 in diabetic nephropathy (DN). Here, mouse mesangial cells (SV40-MES13) were cultured with 30 mM glucose to establish a DN cellular model. The qRT-PCR results indicated that circ_LARP4 expression was downregulated in the DN cellular model compared to that in the control cells. As determined by an MTT assay, circ_LARP4 overexpression via the circ_LARP4 overexpression (OE) plasmids inhibited the cell proliferation rate. As determined by an Annexin V/PI kit and flow cytometry, circ_LARP4 overexpression increased the cell apoptosis rate. As measured by Western blot, circ_LARP4 overexpression enhanced BAX expression but reduced Bcl-2 expression, also suggesting an enhancement of cell apoptosis. Moreover, regarding cell fibrosis, circ_LARP4 overexpression reduced the mRNA levels of fibrosis markers, including fibronectin, collagen I and collagen IV. Interestingly, miR-424 was found to be reduced in the DN cellular model after transfection with the circ_LARP4 OE plasmids. In addition, restoration of miR-424 expression with the miR-424 mimics reversed the negative effects of circ_LARP4 overexpression on cell proliferation and fibrosis. In conclusion, circ_LARP4 was lower in the DN cellular model than in normal cells, and circ_LARP4 overexpression resulted in decreased cell proliferation and cell fibrosis but increased cell apoptosis in the DN cellular model by sponging miR-424.


Subject(s)
DNA, Circular/genetics , Mesangial Cells/metabolism , Proteins/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Diabetic Nephropathies/genetics , Fibrosis , Glucose/metabolism , Mesangial Cells/physiology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , Signal Transduction/genetics
8.
Immunology ; 162(4): 418-433, 2021 04.
Article in English | MEDLINE | ID: mdl-33314123

ABSTRACT

Previously, we demonstrated neuraminidase (NEU) activity or NEU1 expression, specifically, is increased in the kidneys of lupus mice and urine of human patients with nephritis. Additionally, NEU activity mediates IL-6 secretion from lupus-prone MRL/lpr primary mouse mesangial cells (MCs) in response to an IgG mimic. IL-6 mediates glomerular inflammation and promotes tissue damage in patients and mouse strains with lupus nephritis. This study further elucidates the mechanisms by which NEU activity and NEU1 specifically mediates the release of IL-6 and other cytokines from lupus-prone MCs. We demonstrate significantly increased release of multiple cytokines and NEU activity in MRL/lpr MCs in response to serum from MRL/lpr mice (lupus serum). Inhibiting NEU activity significantly reduced secretion of three of those cytokines: IL-6, GM-CSF and MIP1α. Message levels of Il-6 and Gm-csf were also increased in response to lupus serum and reduced when NEU activity was inhibited. Neutralizing antibodies to cell-surface receptors and MAPK inhibitors in lupus serum- or LPS-stimulated MCs indicate TLR4 and p38 or ERK MAP kinase signalling play key roles in the NEU-mediated secretion of IL-6. Significantly reduced IL-6 release was observed in C57BL/6 (B6) Neu1+/+ primary MCs compared with wild-type (Neu1+/+) B6 MCs in response to lupus serum. Additional results show inhibiting NEU activity significantly increases sialic acid-containing N-glycan levels. Together, our novel observations support a role for NEU activity, and specifically NEU1, in mediating release of IL-6 from lupus-prone MCs in response to lupus serum through a TLR4-p38/ERK MAPK signalling pathway that likely includes desialylation of glycoproteins.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/metabolism , Mesangial Cells/physiology , Neuraminidase/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Female , Gene Expression Regulation , Humans , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Neuraminidase/genetics , Serum/metabolism , Signal Transduction
9.
Biochem Biophys Res Commun ; 532(2): 308-314, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32868076

ABSTRACT

BACKGROUND: This study aimed to investigate the involvement of lncRNA CTBP1-AS2 in the progression of diabetic nephropathy (DN) by affecting high glucose (HG)-induced human glomerular mesangial cells (HGMCs). METHODS: HGMCs were selected for the establishment of cell injury induced by HG. The expression of CTBP1-AS2, miR-155-5p and FOXO1 was detected by real-time PCR and western blotting. The target association between miR-155-5p and CTBP1-AS2 or FOXO1 was confirmed by dual-luciferase reporter assays. Cell proliferation and oxidative stress were revealed by CCK-8 colorimetry, and the measurement of reactive oxygen species (ROS) and the activities of antioxidant enzymes. Extracellular matrix (ECM) protein accumulation and the production of inflammatory cytokines were investigated by western blotting and ELISA. RESULTS: The expression of CTBP1-AS2 was downregulated, and miR-155-5p was highly expressed in peripheral blood of DN patients and HG-treated HGMCs. Further investigation revealed that CTBP1-AS2 overexpression inhibited proliferation, oxidative stress, ECM accumulation and inflammatory response in HG-induced HGMCs. Mechanical analysis revealed that CTBP1-AS2 regulated FOXO1 expression via sponging miR-155-5p. Rescue experiments demonstrated that miR-155-5p overexpression or FOXO1 inhibition reversed the effects of CTBP1-AS2 in HG-stimulated HGMCs. CONCLUSION: Taken together, this study revealed CTBP1-AS2 attenuated HG-induced HGMC proliferation, oxidative stress, ECM accumulation, and inflammation through miR-155-5p/FOXO1 signaling.


Subject(s)
Diabetic Nephropathies/genetics , Glomerulonephritis/genetics , MicroRNAs/genetics , RNA, Long Noncoding/blood , Case-Control Studies , Cell Proliferation , Cells, Cultured , Diabetic Nephropathies/blood , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Regulatory Networks , Glomerulonephritis/pathology , Glucose/pharmacology , Humans , Mesangial Cells/drug effects , Mesangial Cells/pathology , Mesangial Cells/physiology , MicroRNAs/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , RNA, Long Noncoding/genetics , Reactive Oxygen Species/metabolism
10.
Toxins (Basel) ; 12(8)2020 08 06.
Article in English | MEDLINE | ID: mdl-32781625

ABSTRACT

The endothelial-to-mesenchymal transition (EndoMT) is involved in the complex pathogenesis of renal fibrosis. The soluble proteoglycan endothelial cell-specific molecule 1 (ESM1) is significantly upregulated in many tumor cells and cirrhosis-related disease. The role of ESM1 in renal fibrosis is unknown. This study investigates the role of ESM1 in renal fibrosis, using an in vivo unilateral ureteral obstruction (UUO) mouse model of renal fibrosis and in vitro mouse kidney MES 13 cells overexpressing ESM1. We observed that ESM1 overexpression significantly increased the motility and migration of MES 13 cells, independent of cell viability. In ESM1-overexpressing MES 13 cells, we also observed elevated expression of mesenchymal markers (N-cadherin, vimentin, matrix metallopeptidase 9 (MMP9)) and the fibrosis marker α-smooth muscle actin (α-SMA) and decreased expression of the endothelial marker vascular endothelial cadherin (VE-cadherin) and CD31. In a mouse model of fibrosis induced by unilateral ureter obstruction, we observed time-dependent increases in ESM1, α-SMA, and vimentin expression and renal interstitial collagen fibers in kidney tissue samples. These results suggest that ESM1 may serve as an EndoMT marker of renal fibrosis progression.


Subject(s)
Kidney Diseases/metabolism , Proteoglycans/physiology , Actins/metabolism , Animals , Cell Line , Cell Movement , Cell Transdifferentiation , Fibrosis , Kidney/metabolism , Kidney/pathology , Kidney Diseases/pathology , Male , Mesangial Cells/physiology , Mice, Inbred C57BL , Vimentin/metabolism
11.
BMC Genomics ; 21(1): 467, 2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32635896

ABSTRACT

BACKGROUND: Mesangial cells play an important role in the glomerulus to provide mechanical support and maintaine efficient ultrafiltration of renal plasma. Loss of mesangial cells due to pathologic conditions may lead to impaired renal function. Mesenchymal stem cells (MSC) can differentiate into many cell types, including mesangial cells. However transcriptomic profiling during MSC differentiation into mesangial cells had not been studied yet. The aim of this study is to examine the pattern of transcriptomic changes during MSC differentiation into mesangial cells, to understand the involvement of transcription factor (TF) along the differentiation process, and finally to elucidate the relationship among TF-TF and TF-key gene or biomarkers during the differentiation of MSC into mesangial cells. RESULTS: Several ascending and descending monotonic key genes were identified by Monotonic Feature Selector. The identified descending monotonic key genes are related to stemness or regulation of cell cycle while ascending monotonic key genes are associated with the functions of mesangial cells. The TFs were arranged in a co-expression network in order of time by Time-Ordered Gene Co-expression Network (TO-GCN) analysis. TO-GCN analysis can classify the differentiation process into three stages: differentiation preparation, differentiation initiation and maturation. Furthermore, it can also explore TF-TF-key genes regulatory relationships in the muscle contraction process. CONCLUSIONS: A systematic analysis for transcriptomic profiling of MSC differentiation into mesangial cells has been established. Key genes or biomarkers, TFs and pathways involved in differentiation of MSC-mesangial cells have been identified and the related biological implications have been discussed. Finally, we further elucidated for the first time the three main stages of mesangial cell differentiation, and the regulatory relationships between TF-TF-key genes involved in the muscle contraction process. Through this study, we have increased fundamental understanding of the gene transcripts during the differentiation of MSC into mesangial cells.


Subject(s)
Cell Differentiation/genetics , Mesangial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Transcriptome , Biomarkers/metabolism , Cells, Cultured , Coculture Techniques , Gene Regulatory Networks , Humans , Mesangial Cells/physiology , Mesenchymal Stem Cells/cytology , Muscle Contraction , Muscle, Smooth, Vascular/physiology , RNA-Seq , Transcription Factors/metabolism
12.
World J Pediatr ; 16(2): 201-212, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31630337

ABSTRACT

BACKGROUND: MicroRNA-29b (miR-29b) has been suggested to possess pro-inflammatory activity, which can partially be explained by the repression of tumor necrosis factor alpha protein three antibody (TNFAIP3). Meanwhile, it also promotes thyroid cell proliferation via Smad signaling pathways. The present study aimed to elucidate the role of miR-29b in Henoch Schönlein purpura nephritis (HSPN) and its underlying molecular mechanism in angiotensin II (Ang II)-induced human glomerular mesangial cell (HGMC) activation. METHODS: We evaluated miR-29b expression in 35 HSPN renal tissues based on crescent formation, glomerular sclerosis, interstitial fibrosis, thrombosis formation and capillary loop necrosis. Meanwhile, HGMCs were cultured, treated with Ang II and then transfected with LV-hsa-miR-29b-1 to induce miR-29b overexpression or LV-hsa-miR-29b-3p-inhibition to inhibit miR-29b expression. Finally, we examined the effects of miR-29b on cell proliferation and release of inflammatory mediators. RESULTS: We observed that miR-29b expression was significantly higher in the crescent group than in the no crescent group. MiR-29b overexpression induced the release of intercellular adhesion molecule-1, interleukin-1ß (IL-1ß), IL-6, IL-8, the increase of CyclinA2, CyclinD1, and cell proliferation. It also could inhibit the expressions of TNFAIP3 and NF-kappa-B-repressing factor (NKRF). Correspondingly, miR-29b inhibition produced the opposite effects and increased the expression of TNFAIP3 and NKRF. CONCLUSION: MiR-29b expression is altered in crescent formation of HSPN and accelerates Ang II-induced mesangial cell proliferation and release of inflammatory mediators.


Subject(s)
Angiotensin II/physiology , Glomerulonephritis/metabolism , IgA Vasculitis/metabolism , Mesangial Cells/physiology , MicroRNAs/biosynthesis , Cell Proliferation , Cells, Cultured , Glomerulonephritis/complications , Humans , IgA Vasculitis/complications , Mesangial Cells/cytology , MicroRNAs/physiology , Time Factors
13.
J Microbiol Immunol Infect ; 53(1): 23-32, 2020 Feb.
Article in English | MEDLINE | ID: mdl-29657028

ABSTRACT

BACKGROUND: Renal disease is prevalent in gouty patients and monosodium urate (MSU) crystal deposition in the kidney can be detected in some gouty nephropathy patients. MSU crystals can induce inflammatory events, we investigated the MSU-induced expression of intercellular adhesion molecule (ICAM)-1 on human renal mesangial cells (HRMCs) and the involved signal transduction mechanisms. METHODS: The HRMCs cell line was purchased from ScienCell Research Laboratories. MSU crystals were made by dissolving uric acid in sodium hydroxide (NaOH) solution. The involvement of MAPKs, apoptosis-associated speck-like protein containing a CARD domain (ASC), and Toll-like receptor (TLR) was investigated using pharmacological inhibitors, transfection with short hairpin RNA (shRNA), or monoclonal antibodies. Protein expression was evaluated by Western blotting. The functional activity of ICAM-1 was evaluated with cell-cell adhesion assay and immunofluorescence analysis. RESULTS: MSU stimulation increased expression of ICAM-1 and adhesion between HRMCs and human monocytic THP-1 cells. The interaction between HRMCs and THP-1 was suppressed by ICAM-1 neutralizing antibodies. MSU stimulation induced activation of mitogen-activated protein kinases, including c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK), but only p38 was responsible for MSU-induced expression of ICAM-1 and cell-cell adhesion. ASC also play a role in MSU-induced effects. Pretreatment with monoclonal antibodies against toll-like receptor (TLR)2 or TLR4 reduced MSU-induced ICAM-1 expression, cell-cell adhesion, p38 phosphorylation but the reduction of ASC activation is insignificant. CONCLUSION: The MSU induced ICAM-1 expression on HRMCs and cell-cell adhesion involved TLR2/4-p38-ICAM1 pathway and TLR2/4 independent ASC-p38-ICAM1 axis. These findings might partly explain the mechanisms underlying gouty nephropathy.


Subject(s)
Cell Adhesion/drug effects , Gout/complications , Intercellular Adhesion Molecule-1/genetics , Kidney Diseases/physiopathology , Mesangial Cells/drug effects , Uric Acid/pharmacology , Cell Line , Humans , Kidney/cytology , Mesangial Cells/physiology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Monocytes/metabolism , Signal Transduction/genetics , THP-1 Cells , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
14.
Biomed Pharmacother ; 121: 109622, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31707340

ABSTRACT

BACKGROUND: Previous study has demonstrated that long noncoding RNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) was abnormally expressed in diabetic nephropathy (DN). However, the underlying mechanism that allows CDKN2B-AS1 in the progression of DN remains to be further elucidated. METHODS: Peripheral blood cells of 24 diabetes patients with DN and 20 without DN were collected. Human glomerular mesangial cells (HGMC) were cultured in high glucose or low glucose medium. The expression levels of CDKN2B-AS1, microRNA (miR)-424-5p and high mobility group AT hook 2 (HMGA2) were detected by quantitative real-time polymerase chain reaction or western blot. The target association between miR-424-5p and CDKN2B-AS1 or HMGA2 was confirmed by dual-luciferase reporter and RNA immunoprecipitation assays. Cell proliferation, extracellular matrix (ECM) accumulation and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling were investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) and western blot, respectively. RESULTS: CDKN2B-AS1 expression was up-regulated and miR-424-5p level was down-regulated in peripheral blood of DN patients and high glucose-treated HGMC cells. CDKN2B-AS1 was validated as a sponge of miR-424-5p. Silence of CDKN2B-AS1 repressed proliferation and ECM accumulation by increasing miR-424-5p. HMGA2 was a target of miR-424-5p and miR-424-5p overexpression inhibited proliferation, ECM accumulation and PI3K/AKT pathway by targeting HMGA2. Moreover, knockdown of CDKN2B-AS1 inhibited HMGA2 expression and PI3K/AKT pathway by increasing miR-424-5p. CONCLUSION: Knockdown of CDKN2B-AS1 suppressed proliferation, ECM accumulation and PI3K/AKT signaling by increasing miR-424-5p and decreasing HMGA2 in high glucose-treated HMGC cells.


Subject(s)
Diabetic Nephropathies/etiology , Extracellular Matrix/metabolism , HMGA2 Protein/physiology , Mesangial Cells/physiology , MicroRNAs/physiology , RNA, Long Noncoding/physiology , Cell Proliferation , Cells, Cultured , Diabetic Nephropathies/metabolism , Humans , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
15.
Biochem Biophys Res Commun ; 520(3): 627-633, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31623827

ABSTRACT

Kidney regenerative medicine is expected to be the solution to the shortage of organs for transplantation. In a previous report, we transplanted exogenous renal progenitor cells (RPCs) including nephron progenitor cells (NPCs), stromal progenitor cells (SPCs), and the ureteric bud (UB) into the nephrogenic zone of animal embryos and succeeded in regenerating new nephrons from exogenous NPCs through a fetal developmental program. However, it was unknown whether the renal stromal lineage cells were regenerated from SPCs. The present study aimed to verify the differentiation of SPCs into mesangial cells and renal stromal lineage cells. Here, we found that simply transplanting RPCs, including SPCs, into the nephrogenic zone of wild-type fetal mice was insufficient for differentiation of SPCs. Therefore, to enrich the purity of SPCs, we sorted cells from RPCs by targeting platelet-derived growth factor receptor alpha (PDGFRa) which is a cell surface marker for immature stromal cells and transplanted the PDGFRa-positive sorted cells. As a result, we succeeded in regenerating a large number of mesangial cells and other renal stromal lineage cells including interstitial fibroblasts, vascular pericytes, and juxtaglomerular cells. We have established the method for regeneration of stromal cells from exogenous SPCs that may contribute to various fields, such as regenerative medicine and kidney embryology, and the creation of disease models for renal stromal disorders.


Subject(s)
Kidney/embryology , Mesangial Cells/physiology , Regeneration/physiology , Animals , Cell Differentiation , Cell Lineage , Female , Green Fluorescent Proteins/genetics , Humans , Kidney/cytology , Kidney/physiology , Male , Mesangial Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Pregnancy , Regenerative Medicine , Stem Cell Transplantation , Stromal Cells/cytology , Stromal Cells/physiology , Stromal Cells/transplantation
16.
Aging (Albany NY) ; 11(20): 8745-8759, 2019 10 20.
Article in English | MEDLINE | ID: mdl-31631065

ABSTRACT

Diabetic nephropathy (DN) is one of the leading causes of end-stage renal diseases worldwide. This study is designed to investigate the underlying function and mechanism of a novel lncRNA GAS5 in the progression of DN. We found that lncRNA GAS5 expression level was decreased in type 2 diabetes (T2D) with DN compared with that in patients without DN. Moreover, lncRNA GAS5 expression level was negatively associated with the severity of DN-related complications. lncRNA GAS5 inhibited MCs proliferation and caused G0/1 phase arrest. lncRNA GAS5 overexpression alleviated the expression of fibrosis-related protein in mesangial cells (MCs). The dual-luciferase reporter assay and RNA binding protein immunoprecipitation (RIP) assay results revealed that lncRNA GAS5 functions as an endogenous sponge for miR-221 via both the directly targeting way and Ago2-dependent manner. Furthermore, SIRT1 was confirmed as a target gene of miR-221. lncRNA GAS5 upregulated SIRT1 expression and inhibited MCs proliferation and fibrosis by acting as an miR-221 sponge. Finally, we found that lncRNA GSA5 suppressed the development of DN in vivo. Thus, lncRNA GAS5 was involved in the progression of DN by sponging miR-221 and contributed to lncRNA-directed diagnostics and therapeutics in DN.


Subject(s)
Diabetic Nephropathies/metabolism , Fibrosis/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Sirtuin 1/metabolism , Aging , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Proliferation , Diabetes Mellitus, Experimental/complications , Gene Deletion , Gene Expression Regulation/drug effects , Glucose/administration & dosage , Glucose/pharmacology , Male , Mesangial Cells/drug effects , Mesangial Cells/physiology , Mice , MicroRNAs/genetics , RAW 264.7 Cells , RNA, Long Noncoding/genetics , Rats , Rats, Sprague-Dawley , Sirtuin 1/genetics
17.
FASEB J ; 33(10): 11492-11506, 2019 10.
Article in English | MEDLINE | ID: mdl-31336052

ABSTRACT

Evidence has shown that long noncoding RNAs (lncRNAs) in the competing endogenous RNA (ceRNA) network are involved in various diseases. However, there is a lack of studies of the ceRNA network in diabetic nephropathy (DN). In this study, we investigated the effect of lncRNAs on mesangial cell (MC) proliferation in DN-related ceRNA networks. Differences in lncRNA and mRNA expression between DN and normal mouse kidney tissues were detected with RNA-seq, and DN-related lncRNA/mRNA/microRNA (miRNA) ceRNA networks were constructed by R3.4.3. Computational analysis was performed, and expression and interactions between the topological RNAs were detected by bioinformatics methods, real-time quantitative PCR (qPCR), and luciferase assay. Cell proliferation ability was measured by 5-ethynyl-2'-deoxyuridine (EdU) in MCs cultured under high- or low-glucose conditions. Moreover, the effect of the topological key lncRNA histocompatibility 2 K region locus 2 (H2k2) H2k2 on MC proliferation via the miRNA (miR)-449a/b/triplet motif 11 (Trim11)/Mek signaling pathway was examined by EdU, flow cytometry analysis, and Western blot. In total, 153 lncRNAs, 428 mRNAs, and 2242 interactions were included in the constructed DN-related ceRNA network. There were 15 RNAs in the top 5% of degree and betweenness. The expression of lncRNA H2k2 and mRNA Trim11 in MCs was increased in DN, which is consistent with the results of RNA-seq and real-time qPCR invivo and in vitro. miR-449a and miR-449b, which were down-regulated in MCs cultured with high glucose, were selected for further analysis. The results of real-time qPCR and luciferase assay revealed the lncRNA H2k2-miR-449a/b-Trim11 interaction in MCs. In addition, the data showed that H2k2 regulates MC proliferation via the miR-449ab/Trim11/Mek signaling pathway. Taken together, these results provide new insight into the association between the topological key lncRNA H2k2 in the DN-related ceRNA network and the miR-449a/b/Trim11/Mek signaling pathway during MC proliferation in DN.-Chen, W., Peng, R., Sun, Y., Liu, H., Zhang, L., Peng, H., Zhang, Z. The topological key lncRNA H2k2 from the ceRNA network promotes mesangial cell proliferation in diabetic nephropathy via the miR-449a/b/Trim11/Mek signaling pathway.


Subject(s)
Cell Proliferation/genetics , Diabetic Nephropathies/genetics , MAP Kinase Signaling System/genetics , Mesangial Cells/physiology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Tripartite Motif Proteins/genetics , Animals , Cell Line , Computational Biology/methods , Gene Regulatory Networks/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , RNA, Messenger/genetics , Signal Transduction/genetics
18.
Int Immunopharmacol ; 70: 313-323, 2019 May.
Article in English | MEDLINE | ID: mdl-30852287

ABSTRACT

Immunoglobulin A nephropathy (IgAN) is an autoimmune kidney disease with complex pathogenesis leading to end-stage renal damage. The crucial pathological characteristic in IgAN is IgA immune complexes deposition accompany with mesangial cell proliferation and mesangial matrix expansion. Artemisinin (ART) is isolated from traditional Chinese medicine Artemisia annua L. Hydroxychloroquine (HCQ) is a classical antimalarial drug used to treat autoimmune diseases. Both of them possess immunosuppressive, immunomodulatory and anti-inflammatory features. The aim of this study was to investigate the pharmacological effects of ART combined with HCQ (AH) and explore the underlying mechanisms in IgAN. In vivo, our results showed that AH could significantly improve kidney dysfunction, decrease mesangial matrix expansion as well as immune complexes in mesangial area visualized by H&E and PAS staining. The depositions of IgA immune complexes and complement 3 (C3) were obviously reduced after AH treatment by immunofluorescence. Interestingly, the morphology of kidney and spleen was significantly swelled but reverted by AH in IgAN rats. Further mechanistic study showed that the higher proportions of the Th2 and Th17 cells were reduced but the lower differentiation of Th1 and Treg cells subsets were promoted by AH. Taken together, this study demonstrated that there was an immunosuppressive effect of AH therapy on IgAN rats via regulating the differentiation of CD4+ T cell subsets, which provided an alternative approach for IgAN treatment.


Subject(s)
Artemisinins/therapeutic use , Drug Therapy, Combination , Glomerulonephritis, IGA/drug therapy , Hydroxychloroquine/therapeutic use , Immunosuppressive Agents/therapeutic use , Mesangial Cells/physiology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Antigen-Antibody Complex/metabolism , Artemisia annua/immunology , CD4 Antigens/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Male , Medicine, Chinese Traditional , Rats , Rats, Sprague-Dawley
19.
Nutrients ; 11(2)2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30744045

ABSTRACT

Perilla frutescens (L.) Britt. var. japonica (Hassk.) Hara (PF), is a medical herb of the Lamiaceae family. We have previously reported that the PF sprout extract (PFSE) is effective in treating hyperglycemia. However, the role of PFSE on glomerular mesangial cells (MCs) proliferation and the extracellular matrix (ECM) accumulation in a diabetic condition are still unclear. Therefore, in this study, we have investigated the role of PFSE on cell proliferation and ECM accumulation in murine glomerular MCs (MMCs), cultured under a high glucose (HG) condition. PFSE treatment attenuated HG-induced MMCs proliferation and hypertrophy. Moreover, the HG-induced ECM protein, collagen IV and fibronectin, overexpression was abolished by the PFFSE treatment. In addition, PFSE inhibited reactive oxygen species (ROS) overproduction and NOX2 and NOX4 expression in MMCs under a HG condition. Our data further revealed the involvement of mesangial cell damage in AMP-activated kinase (AMPK) activation. PFSE strongly activated AMPK in MMCs under hyperglycemic conditions. These results suggest that PFSE inhibits HG-medicated MC fibrosis through suppressing the activation of NOX2/4 and the AMPK activation mechanism. PFSE may be useful for the prevention or treatment of diabetic nephropathy.


Subject(s)
Glucose/adverse effects , Mesangial Cells/drug effects , Perilla frutescens , Plant Extracts/pharmacology , Protective Agents/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Diabetic Nephropathies , Glucose/metabolism , Mesangial Cells/physiology , Mice , NADPH Oxidases/metabolism , Seedlings/chemistry
20.
Acta Pharmacol Sin ; 39(9): 1513-1521, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30150789

ABSTRACT

SND p102 was first described as a transcriptional co-activator, and subsequently determined to be a co-regulator of Pim-1, STAT6 and STAT5. We previously reported that SND p102 expression was increased in high glucose-treated mesangial cells (MCs) and plays a role in the extracellular matrix (ECM) accumulation of MCs by regulating the activation of RAS. In this study, we further examined the roles of SND p102 in diabetic nephropathy (DN)-induced glomerulosclerosis. Rats were injected with STZ (50 mg/kg, ip) to induce diabetes. MCs or isolated glomeruli were cultured in normal glucose (NG, 5.5 mmol/L)- or high glucose (HG, 25 mmol/L)-containing DMEM. We found that SND p102 expression was significantly increased in the diabetic kidneys, as well as in HG-treated isolated glomeruli and MCs. In addition, HG treatment induced significant fibrotic changes in MCs evidenced by enhanced protein expression of TGF-ß, fbronectin and collagen IV, and significantly increased the proliferation of MCs. We further revealed that overexpression of SND p102 significantly increased the protein expression of angiotensin II (Ang II) type 1 receptor (AT1R) in MCs by increasing its mRNA levels via directly targeting the AT1R 3'-UTR, which resulted in activation of the ERK/Smad3 signaling and subsequently promoted the up-regulation of fbronectin, collagen IV, and TGF-ß in MCs, as well as the cell proliferation. These results demonstrate that SND p102 is a key regulator of AT1R-mediating ECM synthesis and cell proliferation in MCs. Thus, small molecule inhibitors of SND p102 may be a novel therapeutic strategy for DN.


Subject(s)
Cell Proliferation/physiology , Diabetic Nephropathies/physiopathology , Extracellular Matrix/metabolism , Kidney/physiopathology , Mesangial Cells/physiology , Nuclear Proteins/metabolism , Animals , Collagen Type IV/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Down-Regulation , Endonucleases , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/metabolism , Fibrosis/physiopathology , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Signaling System/physiology , Male , Nuclear Proteins/genetics , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...