Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 588
Filter
1.
JCI Insight ; 9(9)2024 May 08.
Article in English | MEDLINE | ID: mdl-38716725

ABSTRACT

IgA nephropathy (IgAN) represents the main cause of renal failure, while the precise pathogenetic mechanisms have not been fully determined. Herein, we conducted a cross-species single-cell survey on human IgAN and mouse and rat IgAN models to explore the pathogenic programs. Cross-species single-cell RNA sequencing (scRNA-Seq) revealed that the IgAN mesangial cells (MCs) expressed high levels of inflammatory signatures CXCL12, CCL2, CSF1, and IL-34 and specifically interacted with IgAN macrophages via the CXCL12/CXCR4, CSF1/IL-34/CSF1 receptor, and integrin subunit alpha X/integrin subunit alpha M/complement C3 (C3) axes. IgAN macrophages expressed high levels of CXCR4, PDGFB, triggering receptor expressed on myeloid cells 2, TNF, and C3, and the trajectory analysis suggested that these cells derived from the differentiation of infiltrating blood monocytes. Additionally, protein profiling of 21 progression and 28 nonprogression IgAN samples revealed that proteins CXCL12, C3, mannose receptor C-type 1, and CD163 were negatively correlated with estimated glomerular filtration rate (eGFR) value and poor prognosis (30% eGFR as composite end point). Last, a functional experiment revealed that specific blockade of the Cxcl12/Cxcr4 pathway substantially attenuated the glomerulus and tubule inflammatory injury, fibrosis, and renal function decline in the mouse IgAN model. This study provides insights into IgAN progression and may aid in the refinement of IgAN diagnosis and the optimization of treatment strategies.


Subject(s)
Disease Progression , Glomerulonephritis, IGA , Macrophages , Single-Cell Analysis , Adult , Animals , Female , Humans , Male , Mice , Rats , Chemokine CXCL12/metabolism , Disease Models, Animal , Glomerular Filtration Rate , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/pathology , Interleukins , Macrophages/immunology , Macrophages/metabolism , Mesangial Cells/pathology , Mesangial Cells/metabolism , Mesangial Cells/immunology , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Rats, Wistar
2.
Cell Death Dis ; 15(5): 344, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762508

ABSTRACT

Lupus nephritis (LN) occurs in 50% of cases of systemic lupus erythematosus (SLE) and is one of the most serious complications that can occur during lupus progression. Mesangial cells (MCs) are intrinsic cells in the kidney that can regulate capillary blood flow, phagocytose apoptotic cells, and secrete vasoactive substances and growth factors. Previous studies have shown that various types of inflammatory cells can activate MCs for hyperproliferation, leading to disruption of the filtration barrier and impairment of renal function in LN. Here, we characterized the heterogeneity of kidney cells of LN mice by single-nucleus RNA sequencing (snRNA-seq) and revealed the interaction between macrophages and MCs through the CXC motif chemokine ligand 12 (CXCL12)/dipeptidyl peptidase 4 (DPP4) axis. In culture, macrophages modulated the proliferation and migration of MCs through this ligand-receptor interaction. In LN mice, treatment with linagliptin, a DPP4 inhibitor, effectively inhibited MC proliferation and reduced urinary protein levels. Together, our findings indicated that targeting the CXCL12/DPP4 axis with linagliptin treatment may serve as a novel strategy for the treatment of LN via the CXCL12/DPP4 axis.


Subject(s)
Cell Proliferation , Chemokine CXCL12 , Dipeptidyl Peptidase 4 , Lupus Nephritis , Macrophages , Mesangial Cells , Lupus Nephritis/pathology , Lupus Nephritis/metabolism , Animals , Dipeptidyl Peptidase 4/metabolism , Chemokine CXCL12/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mesangial Cells/drug effects , Mice , Macrophages/metabolism , Cell Proliferation/drug effects , Humans , Female , Cell Movement/drug effects , Cell Communication/drug effects , Linagliptin/pharmacology , Signal Transduction , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Mice, Inbred C57BL
3.
J Diabetes ; 16(6): e13565, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38751373

ABSTRACT

BACKGROUND: Diabetic nephropathy (DN) is a diabetic complication. LncRNAs are reported to participate in the pathophysiology of DN. Here, the function and mechanism of lncRNA small nucleolar RNA host gene 14 (SNHG14) in DN were explored. METHODS: Streptozotocin (STZ)-induced DN mouse models and high glucose (HG)-treated human mesangial cells (MCs) were used to detect SNHG14 expression. SNHG14 silencing plasmids were applied to examine the function of SNHG14 on proliferation and fibrosis in HG-treated MCs. Potential targets of SNHG14 were predicted using bioinformatics tools and verified by luciferase reporter, RNA pulldown, and northern blotting assays. The functional role of SNHG14 in DN in vivo was detected by injection with adenoviral vector carrying sh-SNHG14 into DN mice. Serum creatinine, blood urea nitrogen, blood glucose, 24-h proteinuria, relative kidney weight, and renal pathological changes were examined in DN mice. RESULTS: SNHG14 expression was elevated in the kidneys of DN mice and HG-treated MCs. SNHG14 silencing inhibited proliferation and fibrosis of HG-stimulated MCs. SNHG14 bound to miR-30e-5p to upregulate SOX4 expression. In rescue assays, SOX4 elevation diminished the effects of SNHG14 silencing in HG-treated MCs, and SOX4 silencing reversed the effects of SNHG14 overexpression. In in vivo studies, SNHG14 downregulation significantly ameliorated renal injuries and renal interstitial fibrosis in DN mice. CONCLUSIONS: SNHG14 silencing attenuates kidney injury in DN mice and reduces proliferation and fibrotic phenotype of HG-stimulated MCs via the miR-30e-5p/SOX4 axis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Disease Progression , MicroRNAs , RNA, Long Noncoding , SOXC Transcription Factors , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , RNA, Long Noncoding/genetics , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Mice , MicroRNAs/genetics , Humans , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Male , Gene Silencing , Fibrosis , Cell Proliferation , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice, Inbred C57BL
4.
Toxicol Lett ; 395: 1-10, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38458339

ABSTRACT

The pathogenesis of glomerular diseases is strongly influenced by abnormal extracellular matrix (ECM) deposition in mesangial cells. Dipeptidyl peptidase IV (DPPIV) enzyme family contains DPP8 and DPP9, which are involved in multiple diseases. However, the pathogenic roles of DPP8 and DPP9 in mesangial cells ECM deposition remain unclear. In this study, we observed that DPP8 and DPP9 were significantly increased in glomerular mesangial cells and podocytes in CKD patients compared with healthy individuals, and DPP9 levels were higher in the urine of IgA nephropathy (IgAN) patients than in control urine. Therefore, we further explored the mechanism of DPP8 and DPP9 in mesangial cells and revealed a significant increase in the expression of DPP8 and DPP9 in human mesangial cells (HMCs) following TGF-ß1 stimulation. Silencing DPP8 and DPP9 by siRNAs alleviated the expression of ECM-related proteins including collagen Ⅲ, collagen Ⅳ, fibronectin, MMP2, in TGF-ß1-treated HMCs. Furthermore, DPP8 siRNA and DPP9 siRNA inhibited TGF-ß1-induced phosphorylation of Smad2 and Smad3, as well as the phosphorylation of Akt in HMCs. The findings suggested the inhibition of DPP8/9 may alleviate HMCs ECM deposition induced by TGF-ß1 via suppressing TGF-ß1/Smad and AKT signaling pathways.


Subject(s)
Dipeptidases , Mesangial Cells , Humans , Cells, Cultured , Collagen/metabolism , Dipeptidases/metabolism , Extracellular Matrix/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Signal Transduction , Transforming Growth Factor beta1/metabolism
5.
Cell Commun Signal ; 21(1): 288, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37845726

ABSTRACT

Diabetic kidney disease (DKD) is a major cause of end-stage renal disease and imposes a heavy global economic burden; however, little is known about its complicated pathophysiology. Investigating the cellular crosstalk involved in DKD is a promising avenue for gaining a better understanding of its pathogenesis. Nonetheless, the cellular crosstalk of podocytes and endothelial cells in DKD is better understood than that of mesangial cells (MCs) and renal tubular epithelial cells (TECs). As the significance of MCs and TECs in DKD pathophysiology has recently become more apparent, we reviewed the existing literature on the cellular crosstalk of MCs and TECs in the context of DKD to acquire a comprehensive understanding of their cellular communication. Insights into the complicated mechanisms underlying the pathophysiology of DKD would improve its early detection, care, and prognosis. Video Abstract.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Humans , Diabetic Nephropathies/pathology , Mesangial Cells/pathology , Endothelial Cells/pathology , Epithelial Cells/pathology , Podocytes/pathology , Diabetes Mellitus/pathology
6.
Ann Clin Lab Sci ; 53(2): 192-199, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37094847

ABSTRACT

OBJECTIVE: The dysfunction of mesangial cells is a key contributor to the pathogenesis of diabetic nephropathy, while the underlying molecular basis is not fully elucidated. METHODS: Mouse mesangial cells were administered with high glucose medium and the expression of polo-like kinase 2 (PLK2) was determined by PCR and western blot. Loss-of- and gain-of-function of PLK2 was achieved by small interfering RNA targeting PLK2 or PLK2 overexpression plasmid transfections. The hypertrophy, extracellular matrix production, and oxidative stress of mesangial cells were detected. The activation of p38-MAPK signaling was tested using western blot. SB203580 was employed to block the p38-MAPK signaling. The expression of PLK2 in human renal biopsies was detected by immunohistochemistry. RESULTS: High glucose administration upregulated the expression of PLK2 in mesangial cells. PLK2 knockdown reversed the hypertrophy, extracellular matrix production, and oxidative stress induced by high glucose in mesangial cells. PLK2 knockdown suppressed the activation of p38-MAPK signaling. Blockade of p38-MAPK signaling by SB203580 abolished the dysfunction of mesangial cells induced by high glucose and PLK2 overexpression. The enhanced expression of PLK2 was validated in human renal biopsies. CONCLUSION: PLK2 is a key participant in high glucose-induced mesangial cell dysfunction, and might play a crucial role in the pathogenesis of diabetic nephropathy.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Humans , Mice , Animals , Mesangial Cells/metabolism , Mesangial Cells/pathology , Signal Transduction , Glucose/metabolism , Oxidative Stress , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Hypertrophy/metabolism , Hypertrophy/pathology
7.
Oxid Med Cell Longev ; 2023: 4555609, 2023.
Article in English | MEDLINE | ID: mdl-36718276

ABSTRACT

Methods: The PubMed database was searched to identify all studies related to DN that were published from 2001 to 2021, with these studies being separated into four time-based groups. The characteristics of these studies were analyzed and extracted using BICOMB. Biclustering analyses for each of these groups were then performed using gCLUTO, with these results then being analyzed and GraphPad Prism 5 being used to construct strategy diagrams. The social network analyses (SNAs) for each group of studies were conducted using NetDraw and UCINET. Results: In total, 18,889 DN-associated studies published from 2001 to 2021 and included in the PubMed database were incorporated into the present bibliometric analysis. Biclustering analysis and strategy diagrams revealed that active areas of research interest in the DN field include studies of the drug-based treatment, diagnosis, etiology, pathology, physiopathology, and epidemiology of DN. The specific research topics associated with these individual areas, however, have evolved over time in a dynamic manner. Strategy diagrams and SNA results revealed podocyte metabolism as an emerging research hotspot in the DN research field from 2010 to 2015, while DN-related microRNAs, signal transduction, and mesangial cell metabolism have emerged as more recent research hotspots in the interval from 2016 to 2021. Conclusion: Through analyses of PubMed-indexed studies pertaining to DN published since 2001, the results of this bibliometric analysis offer a knowledge framework and insight into active and historical research hotspots in the DN research space, enabling investigators to readily understand the dynamic evolution of this field over the past two decades. Importantly, these analyses also enable the prediction of future DN-related research hotspots, thereby potentially guiding more focused and impactful research efforts.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , MicroRNAs , Podocytes , Humans , Diabetic Nephropathies/pathology , Podocytes/pathology , Mesangial Cells/pathology , Bibliometrics
8.
Chem Biol Drug Des ; 101(4): 819-828, 2023 04.
Article in English | MEDLINE | ID: mdl-36404132

ABSTRACT

As one of complications of diabetes mellitus, diabetic nephropathy is related to renal dysfunction. Membrane metalloendopeptidase (MME) is associated with the pathogenesis of diabetic nephropathy and exerts a protective function in high glucose (HG)-treated podocytes. Salviolone, one of important bioactive components from Salvia miltiorrhiza, possesses an anti-inflammatory activity. However, the roles of salviolone in renal mesangial cell dysfunction under HG condition remain unknown. The targets of salviolone in diabetic nephropathy were predicted by bioinformatics analysis. Relative mRNA level of MME was detected by qPCR in HG-treated human renal mesangial cells (HRMCs). Cell viability was analyzed using CCK-8 assay. Cell proliferation was investigated by EdU staining. Oxidative stress was evaluated by detection of ROS generation and levels of oxidative stress-related biomarkers. The inflammatory cytokines and fibrosis-related biomarkers were examined by ELISA. Our results showed that MME expression was decreased in diabetic nephropathy and HG-treated HRMCs. Salviolone increased MME level in HG-treated HRMCs. Salviolone mitigated HG-induced HRMC proliferation by increasing MME expression. Salviolone attenuated HG-induced ROS generation, MDA level increase, and SOD activity decrease through upregulating MME expression. Moreover, salviolone suppressed HG-induced increase of levels of TNF-α, IL-1ß, IL-6, fibronectin, and collagen IV through upregulating MME expression. In conclusion, salviolone attenuates proliferation, oxidative stress, inflammation, and fibrosis in HG-treated HRMCs through upregulating MME expression.


Subject(s)
Diabetic Nephropathies , Mesangial Cells , Humans , Cell Proliferation , Fibrosis , Glucose/metabolism , Inflammation/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Neprilysin/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction
9.
Genes (Basel) ; 13(12)2022 11 29.
Article in English | MEDLINE | ID: mdl-36553510

ABSTRACT

Diabetic nephropathy (DN) is a leading cause of end-stage renal disease and continues to be a threat to patients with diabetes. Dysfunction of glomerular mesangial cells (GMCs) is the main contributing factor to glomerulosclerosis, which is a pathological feature of DN. The epigenetic factor ASH2L has long been thought to be a transcriptional activator, but its function and involvement in diabetic nephropathy is still unclear. Here, we investigated the effect of ASH2L on the regulation of fibrosis and inflammation induced by high glucose in mouse mesangial cells (mMCs). We observed that ASH2L expression is increased in high glucose-induced mMCs, while loss of ASH2L alleviated fibrosis and inflammation. Furthermore, ASH2L-mediates H3K4me3 of the homeodomain-interacting protein kinase 2 (HIPK2) promoter region, which is a contributor to fibrosis in the kidneys and promotes its transcriptional expression. Similar to loss of ASH2L, silencing HIPK2 also inhibited fibrosis and inflammation. In addition, ASH2L and HIPK2 are upregulated in the kidneys of both streptozocin-induced and db/db mouse. In conclusion, we uncovered the crucial role of ASH2L in high glucose-induced fibrosis and inflammation, suggesting that ASH2L regulation may be an attractive approach to attenuate the progression of DN.


Subject(s)
DNA-Binding Proteins , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Transcription Factors , Animals , Mice , Diabetes Mellitus, Experimental/chemically induced , Diabetic Nephropathies/etiology , Fibrosis , Glucose/toxicity , Inflammation/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism
10.
Oxid Med Cell Longev ; 2022: 1740770, 2022.
Article in English | MEDLINE | ID: mdl-36388165

ABSTRACT

Growing evidence suggests that mesangial cells (MCs) play a crucial role in the pathogenesis of IgA nephropathy (IgAN) by secreting aIgA1. However, the mechanism by which MCs regulate podocyte injury remains unknown. This study demonstrated that MC-derived exosomes treated with aIgA1 induced podocyte injury in IgA nephropathy. miR-4455, which was significantly upregulated in aIgA1 treatment MC-derived exosomes, can be transferred from MCs to podocytes via exosomes. MC-derived exosomal miR-4455 induced podocyte injury. Mechanistically, exosomal miR-4455 directly targeted ULK2 to regulate LC3II/I and P62 levels, which mediates autophagy homeostasis. This study revealed that MC-derived exosomal miR-4455 is a key factor affecting podocyte injury and provides a series of potential therapeutic targets for treating IgA nephropathy.


Subject(s)
Glomerulonephritis, IGA , MicroRNAs , Podocytes , Humans , Glomerulonephritis, IGA/genetics , Glomerulonephritis, IGA/pathology , Mesangial Cells/pathology , Podocytes/pathology , Autophagy/genetics , MicroRNAs/genetics , Protein Serine-Threonine Kinases
11.
J Biomed Sci ; 29(1): 31, 2022 May 10.
Article in English | MEDLINE | ID: mdl-35538534

ABSTRACT

BACKGROUND: Mesangial cell fibrosis, a typical symptom of diabetic nephropathy (DN), is a major contributor to glomerulosclerosis. We previously reported that the pharmacological blockade of lysophosphatidic acid (LPA) signaling improves DN. Although LPA signaling is implicated in diabetic renal fibrosis, the underlying molecular mechanisms remain unclear. Here, the role of carbohydrate-responsive element-binding protein (ChREBP) in LPA-induced renal fibrosis and the underlying mechanisms were investigated. METHODS: Eight-week-old wild-type and db/db mice were intraperitoneally injected with the vehicle or an LPAR1/3 antagonist, ki16425 (10 mg/kg), for 8 weeks on a daily basis, following which the mice were sacrificed and renal protein expression was analyzed. SV40 MES13 cells were treated with LPA in the presence or absence of ki16425, and the expression of ChREBP and fibrotic factors, including fibronectin, TGF-ß, and IL-1ß, was examined. The role of ChREBP in the LPA-induced fibrotic response was investigated by ChREBP overexpression or knockdown. The involvement of Smad ubiquitination regulatory factor-2 (Smurf2), an E3 ligase, in LPA-induced expression of ChREBP and fibrotic factors was investigated by Smurf2 overexpression or knockdown. To identify signaling molecules regulating Smurf2 expression by LPA, pharmacological inhibitors such as A6370 (Akt1/2 kinase inhibitor) and Ly 294002 (PI3K inhibitor) were used. RESULTS: The renal expression of ChREBP increased in diabetic db/db mice, and was reduced following treatment with the ki16425. Treatment with LPA induced the expression of ChREBP and fibrotic factors, including fibronectin, TGF-ß, and IL-1ß, in SV40 MES13 cells, which were positively correlated. The LPA-induced expression of fibrotic factors increased or decreased following ChREBP overexpression and knockdown, respectively. The production of reactive oxygen species (ROS) mediated the LPA-induced expression of ChREBP and fibrotic factors, and LPA decreased Smurf2 expression via Traf4-mediated ubiquitination. The LPA-induced expression of ubiquitinated-ChREBP increased or decreased following Smurf2 overexpression and knockdown, respectively. Additionally, Smurf2 knockdown significantly increased the expression of ChREBP and fibrotic factors. The pharmacological inhibition of Akt signaling suppressed the LPA-induced alterations in the expression of ChREBP and Smurf2. CONCLUSION: Collectively, the results demonstrated that the ROS/Akt-dependent downregulation of Smurf2 and the subsequent increase in ChREBP expression might be one of the mechanisms by which LPA induces mesangial cell fibrosis in DN.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Diabetic Nephropathies , Lysophospholipids , Mesangial Cells , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Ubiquitin-Protein Ligases , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Down-Regulation , Female , Fibronectins/metabolism , Fibrosis , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TNF Receptor-Associated Factor 4/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Nephrology (Carlton) ; 27(5): 458-466, 2022 May.
Article in English | MEDLINE | ID: mdl-35213087

ABSTRACT

AIM: Tenascin-C (TNC), a non-structural extracellular matrix glycoprotein, is transiently expressed during development or after injury, playing an important role in injury and repair process. The potential role of TNC in the pathogenesis of IgA nephropathy (IgAN) remains to be clarified. METHODS: Immunohistochemistry staining for TNC was conducted on paraffin-embedded slices from renal biopsies of 107 IgAN patients, and correlation analysis was made between mesangial TNC expression and clinic-pathological parameters. In situ hybridization for TNC mRNA was further performed to figure out the cells that express TNC within glomeruli. In vitro experiments were also carried out on mouse mesangial cells (SV40 MES13) to elucidate the effect of TNC on mesangial cells. RESULTS: TNC was expressed in the mesangial area of IgAN, as well as in fibrotic regions. Correlation analysis showed that higher mesangial TNC was associated with higher level of proteinuria, lower estimated glomerular filtration rate and more serious pathological lesions (MEST score). In situ hybridization revealed that abundant TNC mRNA expression was observed in the affected glomeruli of IgAN, but not in minimal change disease. Moreover, TNC mRNA co-localized with PDGFRß mRNA, but not with PODXL mRNA, suggesting that TNC mRNA was expressed in the mesangial cells within glomeruli in IgAN. In vitro experiments showed that exogenous TNC promoted matrix protein production and mesangial cell proliferation, which was attenuated by an epidermal growth factor receptor inhibitor. CONCLUSION: Taken together, these results suggest that mesangial cell-derived TNC contributes to mesangial matrix expansion and mesangial cell proliferation, which is a potential therapeutic target in IgAN.


Subject(s)
Glomerulonephritis, IGA , Mesangial Cells , Animals , Cell Proliferation , Extracellular Matrix/metabolism , Glomerulonephritis, IGA/pathology , Humans , Mesangial Cells/pathology , Mice , Tenascin/genetics , Tenascin/pharmacology
13.
IUBMB Life ; 74(5): 419-432, 2022 05.
Article in English | MEDLINE | ID: mdl-35103378

ABSTRACT

Our previous study showed that ENSMUST00000147869 was abnormally low expressed in the early stage of diabetic nephropathy (DN). ENSMUST00000147869 could inhibit the fibrosis and proliferation of mouse mesangial cells (MMCs), but the mechanism is still unclear. This study aims to explore the specific mechanism underline ENSMUST00000147869 regulates the proliferation and fibrosis of MMCs in DN. Nucleocytoplasmic fractionation was applied to define the location of ENSMUST00000147869 in MMCs. RNA-protein pulldown, RNA immunoprecipitation and mass spectrometry were used to identify upregulated Hspa9 directly interacting with ENSMUST00000147869. SiRNA and lentivirus packaging were used to clarify the role of Hspa9 downregulated by ENSMUST00000147869 in promoting proliferation and fibrosis in MMCs. CHX and MG132 were used to clarify the regulatory role of ENSMUST00000147869 to Hspa9. Immunoprecipitation confirmed the binding of Hspa9 and HMGB1. HSPA9 was a direct binding protein of ENSMUST00000147869, and ENSMUST00000147869 could inhibit proliferation and fibrosis of MMCs by down-regulating HSPA9 through ubiquitination process. HMGB1 was the downstream binding protein of Hspa9, and ENSMUST00000147869 could inhibit the interaction between Hspa9 and HMGB1. Our data showed that ENSMUST00000147869 regulates Hspa9 through the ubiquitin proteasome pathway and inhibits the binding of Hspa9 and HMGB1. The ENSMUST00000147869/Hspa9/HMGB1 axis may act as a diagnostic molecular marker and an effective therapeutic target for DN.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , HMGB1 Protein , Animals , Cell Proliferation/genetics , Diabetes Mellitus/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Female , Fibrosis , HMGB1 Protein/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Male , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mitochondrial Proteins/metabolism , RNA, Small Interfering/metabolism
14.
Curr Rheumatol Rep ; 23(12): 83, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34985599

ABSTRACT

PURPOSE OF REVIEW: Mesangial cells are critical for the proper function of the glomerulus, playing roles in structural support and injury repair. However, they are also early responders to glomerular immune complex deposition and contribute to inflammation and fibrosis in lupus nephritis. This review highlights recent studies identifying signaling pathways and mediators in mesangial cell response to lupus-relevant stimuli. RECENT FINDINGS: Anti-dsDNA antibodies, serum, or plasma from individuals with lupus nephritis, or specific pathologic factors activated multiple signaling pathways. These pathways largely included JAK/STAT/SOCS, PI3K/AKT, and MAPK and led to induction of proliferation and expression of multiple proinflammatory cytokines, growth factors, and profibrotic factors. NFκB activation was a common mediator of response. Mesangial cells proliferate and express a wide array of proinflammatory/profibrotic factors in response to a variety of lupus-relevant pathologic stimuli. While some of the responses are similar, the mechanisms involved appear to be diverse depending on the stimulus. Future studies are needed to fully elucidate these mechanisms with respect to the diverse milieu of stimuli.


Subject(s)
Lupus Nephritis , Mesangial Cells , Antibodies, Antinuclear , Fibrosis , Humans , Lupus Nephritis/pathology , Mesangial Cells/pathology , Phosphatidylinositol 3-Kinases
15.
Biochem Genet ; 60(5): 1471-1487, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34731387

ABSTRACT

Diabetic nephropathy (DN) is the main cause of end-stage renal disease. Circular RNA hsa_circ_0004442 (circTLK1) accelerates the progression of renal cell carcinoma. However, the role of circTLK1 in DN pathogenesis is indistinct. The expression of circTLK1, microRNA-126-5p (miR-126-5p), and microRNA-204-5p (miR-204-5p) was tested by quantitative real-time polymerase chain reaction. The levels of interleukin-6 and interleukin-1ß were measured by enzyme-linked immunosorbent assay. The levels of reactive oxygen species and malondialdehyde and the activity of superoxide dismutase were determined with corresponding kits. Several protein levels were evaluated with western blotting. The relationship between circTLK1 and miR-126-5p/miR-204-5p was verified by dual-luciferase reporter assay. CircTLK1 was highly expressed in DN patient's serum and high-glucose (HG)-treated human mesangial cells. Functionally, circTLK1 inhibition reduced HG-induced inflammation, oxidative stress, and ECM accumulation in human mesangial cells. CircTLK1 was verified as a sponge for miR-126-5p and miR-204-5p, which were downregulated in DN patient's serum and HG-treated human mesangial cells. Both miR-126-5p and miR-204-5p upregulation decreased inflammation, oxidative stress, and ECM accumulation in HG-treated human mesangial cells and circTLK1 silencing-mediated influence on HG-induced human mesangial cell injury was overturned by miR-126-5p or miR-204-5p inhibition. Moreover, circTLK1 knockdown blocked the AKT/NF-κB pathway by sponging miR-126-5p/miR-204-5p. CircTLK1 downregulation alleviated HG-induced inflammation, oxidative stress, and ECM accumulation through blocking the AKT/NF-κB pathway via sponging miR-126-5p/miR-204-5p, providing a new mechanism to comprehend the pathogenesis of DN.


Subject(s)
Diabetic Nephropathies , MicroRNAs , Protein Serine-Threonine Kinases , RNA, Circular , Cell Proliferation/physiology , Diabetic Nephropathies/genetics , Down-Regulation , Glucose/pharmacology , Humans , Inflammation/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Circular/genetics
16.
Int J Mol Med ; 49(1)2022 01.
Article in English | MEDLINE | ID: mdl-34779492

ABSTRACT

Glomerular mesangial cell (MC) proliferation and extracellular matrix deposition are the main pathological changes in diabetic nephropathy. Hydrogen sulfide (H2S) inhibits the proliferation of MCs. Dopamine 1 receptors (DR1) are expressed in MCs and serve important physiological roles. However, it is unclear whether DR1 activation inhibits MC proliferation by increasing endogenous H2S. The present study found that the production of H2S and the expression of DR1 and cystathionine­Î³­lyase (CSE) were decreased in the renal tissues of diabetic mice and high glucose (HG)­induced MCs. SKF38393 (a DR1 agonist) increased the production of H2S and the expression of DR1 and CSE and NaHS (an exogenous H2S donor) only increased H2S production and CSE expression but not DR1 expression. HG increased the thickness of the glomerular basement membrane, cell viability and proliferation, the expression of cyclin D1, PCNA, collagen 1 and α­smooth muscle actin and the activity of phosphorylated ERK1/2 and decreased the expression of P21 and MMP9. SKF38393 and NaHS reversed the effects of HG. PPG (a CSE inhibitor) abolished the beneficial effects of SKF38393. The beneficial effects of SKF38393 were similar to those of PD98059 (an ERK1/2 inhibitor). Taken together, the findings suggested that the DR1­CSE/H2S pathway activation attenuated diabetic MC proliferation and extracellular matrix deposition by downregulating the ERK1/2 signaling pathway.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Diabetes Mellitus, Experimental/pathology , Hydrogen Sulfide/metabolism , Kidney/pathology , Receptors, Dopamine D1/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Cell Line , Cell Proliferation , Collagen/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Fibrosis , Glucose/pharmacology , Kidney/metabolism , MAP Kinase Signaling System/physiology , Male , Mesangial Cells/drug effects , Mesangial Cells/pathology , Mice, Inbred C57BL , Receptors, Dopamine D1/agonists
17.
Dis Markers ; 2021: 3803601, 2021.
Article in English | MEDLINE | ID: mdl-34820026

ABSTRACT

OBJECTIVES: Most patients with systemic lupus erythematosus (SLE) develop lupus nephritis (LN) with severe kidney manifestations. Renal fibrosis can be primarily attributed to overproliferation of mesangial cells (MCs), which are subject to drug treatment. Nevertheless, the detailed mechanisms remain elusive. We sought to identify the effect of cyclophosphamide (CTX), a drug commonly used for LN treatment, on MC proliferation and explore its underlying mechanisms. Material/Methods. Cell proliferation and fibrosis in mouse kidney tissues were determined by histopathology staining techniques. Flow cytometry was used for cell cycle analysis. Cell cycle regulators were examined in vitro following treatment of immortalized human MCs with platelet-derived growth factor subunit B (PDGF-B). Quantitative real-time PCR and western blot analyses were used to measure the mRNA and protein levels of candidate cell cycle regulators, respectively. RESULTS: CTX inhibited cell overproliferation induced by platelet-derived growth factor subunit B in vitro and in vivo. CTX (40 mg/l) was sufficient to induce G0/G1 phase cell cycle arrest. CTX treatment downregulated many critical cell cycle regulators including cyclins and cyclin-dependent kinases but upregulated cyclin-dependent kinase inhibitors. Additionally, CTX-treated samples showed significantly reduced fibrosis, as indicated by lower expression of interleukin-1ß and α-smooth muscle actin. CONCLUSION: CTX inhibits proliferation of MCs by modulating cell cycle regulator and therefore arresting them at G1 phase. CTX treatment significantly alleviates the severity of renal fibrosis. These findings provide novel insights into the mechanisms by which CTX affects LN.


Subject(s)
Cell Cycle Checkpoints , Cyclophosphamide/pharmacology , Fibrosis/drug therapy , Immunosuppressive Agents/pharmacology , Lupus Nephritis/complications , Mesangial Cells/drug effects , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Female , Fibrosis/etiology , Fibrosis/pathology , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL
18.
Eur J Pharmacol ; 913: 174629, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34780751

ABSTRACT

Diabetic nephropathy (DN) is one of the major microvascular complications of diabetes. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial cellular defense factor to cope with oxidative stress. Silent information regulator T1 (Sirt1) is a deacetylase with antioxidative stress activity. Fucoxanthin is a marine-derived carotenoid. This study was conducted to investigate whether fucoxanthin could alleviate oxidative stress by activating Sirt1/Nrf2 signaling to alleviate DN. In streptozotocin-induced diabetic rats, fucoxanthin treatment effectively improved renal function, alleviated glomerulosclerosis. Fucoxanthin reversed the decreased protein levels of Sirt1 and Nrf2 in the kidney of diabetic rats and glomerular mesangial cells cultured in high glucose. Conversely, EX527, a Sirt1 inhibitor, counteracted the effect of fucoxanthin on the expression of Nrf2. Furthermore, in vivo and vitro results showed that fucoxanthin treatment reversed the low expression and activity of superoxide dismutase and heme oxygenase 1, depending on Sirt1 activation. Our results suggest that fucoxanthin improves diabetic kidney function and renal fibrosis by activating Sirt1/Nrf2 signaling to reduce oxidative stress.


Subject(s)
Antioxidants/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/drug therapy , Mesangial Cells/pathology , Xanthophylls/pharmacology , Animals , Antioxidants/therapeutic use , Carbazoles/pharmacology , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Fibrosis , Heme Oxygenase (Decyclizing)/metabolism , Humans , Male , Mesangial Cells/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Streptozocin/administration & dosage , Streptozocin/toxicity , Xanthophylls/therapeutic use
19.
Int J Biol Sci ; 17(13): 3343-3355, 2021.
Article in English | MEDLINE | ID: mdl-34512151

ABSTRACT

Mesangial cell (MC) proliferation and matrix expansion are basic pathological characteristics of IgA nephropathy (IgAN). However, the stepwise mechanism of MC proliferation and the exact set of related signaling molecules remain largely unclear. In this study, we found a significant upregulation of miR-214-3p in the renal cortex of IgAN mice by miRNA sequencing. In situ hybridization analysis showed that miR-214-3p expression was obviously elevated in MCs in the renal cortex in IgAN. Functionally, knockdown of miR-214-3p alleviated mesangial hypercellularity and renal lesions in IgAN mice. In vitro, the inhibition of miR-214-3p suppressed MC proliferation and arrested G1-S cell cycle pSrogression in IgAN. Mechanistically, a luciferase reporter assay verified PTEN as a direct target of miR-214-3p. Downregulation of miR-214-3p increased PTEN expression and reduced p-JNK and p-c-Jun levels, thereby inhibiting MC proliferation and ameliorating renal lesions in IgAN. Moreover, these changes could be attenuated by co-transfection with PTEN siRNA. Collectively, these results illustrated that miR-214-3p accelerated MC proliferation in IgAN by directly targeting PTEN to modulate JNK/c-Jun signaling. Therefore, miR-214-3p may represent a novel therapeutic target for IgAN.


Subject(s)
Glomerulonephritis, IGA/metabolism , Mesangial Cells/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Female , Glomerulonephritis, IGA/pathology , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Mesangial Cells/pathology , Mice, Inbred BALB C , Proto-Oncogene Proteins c-jun/metabolism , Random Allocation
20.
Front Endocrinol (Lausanne) ; 12: 690784, 2021.
Article in English | MEDLINE | ID: mdl-34512545

ABSTRACT

Diabetic nephropathy (DN) is a serious complication of diabetes mellitus. Long non-coding RNAs (lncRNAs) are regulators in DN progression. However, the regulatory mechanisms of multiple lncRNAs in DN remain to be determined. Our aim was to investigate the function and molecular mechanism of lncRNA RNA component of mitochondrial RNAase P (Rmrp) in DN. Here, we observed that the expression of Rmrp was up-regulated in the kidney of db/db DN mice and high glucose induced glomerular mesangial cells (MC). More importantly, the abnormal transcription of Rmrp was induced by nuclear transcription factor Sp1, which promotes the proliferation and production of fibrotic markers in MC. Subsequently, we screened the miRNAs related to Rmrp and found that Rmrp and miR-1a-3p are co-localized at the subcellular level of MC, and Rmrp could directly binds to miR-1a-3p. Further mechanism research demonstrated that the elevated miR-1a-3p significantly attenuated the proliferation and fibrosis-promoting effects induced by up-regulation of Rmrp. At the same time, we also investigated that miR-1a-3p can directly bind to Jun D proto-oncogene (JunD), thereby regulating the protein level of JunD. Rmrp-induced proliferation and fibrogenesis were reversed by co-transfection with JunD siRNA. In summary, Sp1 induced lncRNA Rmrp could drive the expression of JunD via sponging miR-1a-3p in DN progression.


Subject(s)
Cell Proliferation/genetics , Diabetic Nephropathies/pathology , Mesangial Cells/pathology , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Sp1 Transcription Factor/metabolism , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Kidney/metabolism , Kidney/pathology , Male , Mesangial Cells/metabolism , Mice , MicroRNAs/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Long Noncoding/genetics , Sp1 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...