Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.154
Filter
1.
J Vis Exp ; (210)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39248530

ABSTRACT

Membranous nephropathy (MN) is a common pathological type of adult nephrotic syndrome. Up to 20% of patients with MN develop end-stage renal disease (ESRD). Podocytes have an important function in maintaining the glomerular filtration barrier and play a crucial role in the occurrence and development of proteinuria and MN. PI3K/AKT signaling pathway is involved in the entire process of podocyte growth, differentiation, and apoptosis. Kemeng Fang (KMF) is a traditional Chinese medicine formula that has been used to delay kidney injury. However, the therapeutic mechanism of KMF in MN is unclear. Here, the MN rat model was established by axillary, inguinal, and tail vein injections of cationized bovine serum albumin (C-BSA), and then KMF and PI3K inhibitor (LY294002) were administered. The data of liver function, kidney function, blood lipid, renal pathology, podocyte function, expression level of PI3K/AKT signaling pathway, and transcriptomics of rats demonstrated that KMF has a protective effect on the podocytes of MN rats by activating the PI3K/AKT signaling pathway, and it can effectively prevent the progression of MN.


Subject(s)
Apoptosis , Drugs, Chinese Herbal , Glomerulonephritis, Membranous , Phosphatidylinositol 3-Kinases , Podocytes , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Glomerulonephritis, Membranous/pathology , Glomerulonephritis, Membranous/drug therapy , Glomerulonephritis, Membranous/metabolism , Podocytes/drug effects , Podocytes/metabolism , Podocytes/pathology , Rats , Signal Transduction/drug effects , Apoptosis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Drugs, Chinese Herbal/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Male , Rats, Sprague-Dawley , Morpholines/pharmacology , Morpholines/therapeutic use , Chromones/pharmacology , Disease Models, Animal
2.
Cells ; 13(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273018

ABSTRACT

Peroxisome proliferator-activated receptor-alpha (PPAR-α) and its exogenous activators (fibrates) promote autophagy. However, whether the deleterious effects of PPAR-α deficiency on doxorubicin (DOX)-induced podocytopathy are associated with reduced autophagy remains to be clarified. We investigated the mechanisms of PPAR-α in DOX-induced podocytopathy and tubular injury in PPAR-α knockout (PAKO) mice and in a murine podocyte cell line. DOX-treated PAKO mice showed higher serum levels of triglycerides and non-esterified fatty acids and more severe podocytopathy than DOX-treated wild-type mice, as evidenced by higher urinary levels of proteins and podocalyxin at 3 days to 2 weeks and higher blood urea nitrogen and serum creatinine levels at 4 weeks. Additionally, there was an increased accumulation of p62, a negative autophagy marker, in the glomerular and tubular regions in DOX-treated PAKO mice at Day 9. Moreover, DOX-treated PAKO mice showed more severe glomerulosclerosis and tubular damage and lower podocalyxin expression in the kidneys than DOX-treated control mice at 4 weeks. Furthermore, DOX treatment increased p-p53, an apoptosis marker, and cleaved the caspase-3 levels and induced apoptosis, which was ameliorated by fenofibrate, a PPAR-α activator. Fenofibrate further enhanced AMPK activation and autophagy under fed and fasting conditions. Conclusively, PPAR-α deficiency enhances DOX-induced podocytopathy, glomerulosclerosis, and tubular injury, possibly by reducing autophagic activity in mouse kidneys.


Subject(s)
Autophagy , Doxorubicin , Mice, Knockout , PPAR alpha , Podocytes , Animals , Podocytes/metabolism , Podocytes/pathology , Podocytes/drug effects , Doxorubicin/adverse effects , PPAR alpha/metabolism , PPAR alpha/genetics , Mice , Autophagy/drug effects , Cell Line , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Diseases/metabolism , Kidney Diseases/genetics , Apoptosis/drug effects , Fenofibrate/pharmacology , Male
3.
Mol Biol Rep ; 51(1): 1003, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39305364

ABSTRACT

BACKGROUND: Poricoic acid A (PAA), a major triterpenoid component of Poria cocos with anti-tumor, anti-fibrotic, anti-inflammatory, and immune-regulating activities, has been shown to induce podocyte autophagy in diabetic kidney disease (DKD) by downregulating FUN14 domain containing 1 (FUNDC1). This study aimed to identify the role of adenosine monophosphate-activated protein kinase alpha (AMPKα) in PAA-mediated phosphorylation of FUNDC1 in podocyte injury occurring in the pathogenesis of DKD. METHODS AND RESULTS: A cellular model of renal podocyte injury was established by culturing MPC5 cells under high-glucose (HG) conditions. MPC5 cells were subjected to transfection with small interfering RNA (siRNA) targeting AMPKα or siRNA targeting FUNDC1, an AMPKα activator, or PAA. PAA treatment induced the phosphorylation of AMPKα in HG-cultured podocytes. AMPKα activation was implicated in the inhibitory effect of PAA on FUNDC phosphorylation in HG-cultured podocytes. Treatment targeting the AMPKα activator also significantly augmented proliferation, migration, mitochondrial membrane potential, and autophagy levels, while reducing apoptosis levels, inhibiting oxidative stress, and suppressing the release of proinflammatory factors in HG-cultured MPC5 cells. In contrast, insufficient expression of AMPKα reversed the effects of PAA on the proliferation, migration, and apoptosis of podocytes and further exacerbated the reduction of phosphorylated FUNDC1 expression in podocytes under HG conditions. CONCLUSIONS: AMPKα is involved in the regulation of FUNDC1 phosphorylation by PAA in HG-induced podocyte injury. Furthermore, the AMPKα/FUNDC1 pathway plays a crucial regulatory role in HG-induced podocyte injury. These findings support AMPKα, FUNDC1, and the AMPKα/FUNDC1 pathway as targets for PAA intervention.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Diabetic Nephropathies , Glucose , Membrane Proteins , Podocytes , Signal Transduction , Triterpenes , Podocytes/drug effects , Podocytes/metabolism , Podocytes/pathology , AMP-Activated Protein Kinases/metabolism , Animals , Signal Transduction/drug effects , Mice , Autophagy/drug effects , Phosphorylation/drug effects , Membrane Proteins/metabolism , Triterpenes/pharmacology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Oxidative Stress/drug effects , Membrane Potential, Mitochondrial/drug effects
4.
Nat Commun ; 15(1): 8038, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271683

ABSTRACT

Diabetic kidney disease (DKD) is the main cause of chronic kidney disease worldwide. While injury to the podocytes, visceral epithelial cells that comprise the glomerular filtration barrier, drives albuminuria, proximal tubule (PT) dysfunction is the critical mediator of DKD progression. Here, we report that the podocyte-specific induction of human KLF6, a zinc-finger binding transcription factor, attenuates podocyte loss, PT dysfunction, and eventual interstitial fibrosis in a male murine model of DKD. Utilizing combination of snRNA-seq, snATAC-seq, and tandem mass spectrometry, we demonstrate that podocyte-specific KLF6 triggers the release of secretory ApoJ to activate calcium/calmodulin dependent protein kinase 1D (CaMK1D) signaling in neighboring PT cells. CaMK1D is enriched in the first segment of the PT, proximal to the podocytes, and is critical to attenuating mitochondrial fission and restoring mitochondrial function under diabetic conditions. Targeting podocyte-PT signaling by enhancing ApoJ-CaMK1D might be a key therapeutic strategy in attenuating the progression of DKD.


Subject(s)
Diabetic Nephropathies , Kidney Tubules, Proximal , Kruppel-Like Factor 6 , Podocytes , Signal Transduction , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/genetics , Podocytes/metabolism , Podocytes/pathology , Animals , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Humans , Mice , Kruppel-Like Factor 6/metabolism , Kruppel-Like Factor 6/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Mice, Inbred C57BL , Disease Models, Animal
5.
Int J Biol Sci ; 20(12): 4654-4673, 2024.
Article in English | MEDLINE | ID: mdl-39309435

ABSTRACT

Diabetic kidney disease (DKD) is becoming the most leading cause of end-stage renal disease (ESRD). Podocyte injury plays a critical role in DKD progression. Notably, mitochondrial dysfunction is crucial for podocyte injury. MicroRNAs (miRNAs) involves in various kidney diseases. Herein, we discovered miR-29b was induced in the urine of 126 patients with DKD (stage I and II), and negatively correlated with kidney function and podocyte homeostasis. Mechanically, miR-29b targeted peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a co-activator of transcription factors regulating mitochondrial biogenesis and energy metabolism. In vitro, ectopic miR-29b downregulated PGC-1α and promoted podocyte injury, while inhibition of miR-29b alleviated podocyte injury. Consistently, inhibition of miR-29b mitigated podocyte injury and preserved kidney function in ADR nephropathy and db/db mice, and overexpression of miR-29b accelerated disease. Knockout miR-29b specifically in podocyte inhibited mitochondrial dysfunction and podocyte injury. These results revealed miR-29b plays a crucial role in mitochondrial dysfunction through targeted inhibition on PGC-1α, leading to podocyte injury and DKD progression. Importantly, miR-29b could serve as a novel biomarker of podocyte injury and assists to early diagnose DKD.


Subject(s)
Diabetic Nephropathies , MicroRNAs , Mitochondria , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Podocytes , Podocytes/metabolism , Podocytes/pathology , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Mice , Mitochondria/metabolism , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Male , Mice, Inbred C57BL , Female
6.
Cell Death Dis ; 15(9): 697, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349450

ABSTRACT

Mitochondrial dysfunction is a significant contributor to podocyte injury in diabetic kidney disease (DKD). While previous studies have shown that PVT1 might play a vital role in DKD, the precise molecular mechanisms are largely unknown. By analyzing the plasma and kidney tissues of DKD patients, we observed a significant upregulation of PVT1 expression, which exhibited a positive correlation with albumin/creatinine ratios and serum creatinine levels. Then, we generated mice with podocyte-specific deletion of PVT1 (Nphs2-Cre/Pvt1flox/flox) and confirmed that the deletion of PVT1 suppressed podocyte mitochondrial dysfunction and inflammation in addition to ameliorating diabetes-induced podocyte injury, glomerulopathy, and proteinuria. Subsequently, we cultured podocytes in vitro and observed that PVT1 expression was upregulated under hyperglycemic conditions. Mechanistically, we demonstrated that PVT1 was involved in mitochondrial dysfunction by interacting with TRIM56 post-transcriptionally to modulate the ubiquitination of AMPKα, leading to aberrant mitochondrial biogenesis and fission. Additionally, the release of mtDNA and mtROS from damaged mitochondria triggered inflammation in podocytes. Subsequently, we verified the important role of TRIM56 in vivo by constructing Nphs2-Cre/Trim56flox/flox mice, consistently with the results of Nphs2-Cre/Pvt1flox/flox mice. Together, our results revealed that upregulation of PVT1 could promote mitochondrial dysfunction and inflammation of podocyte by modulating TRIM56, highlighting a potential novel therapeutic target for DKD.


Subject(s)
Diabetic Nephropathies , Mitochondria , Podocytes , RNA, Long Noncoding , Podocytes/metabolism , Podocytes/pathology , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mitochondria/metabolism , Mice , Humans , Male , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Mice, Inbred C57BL , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Female
7.
Ren Fail ; 46(2): 2394637, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39189638

ABSTRACT

BACKGROUND: Recent studies have reported that helix B surface polypeptide (HBSP), an erythropoietin derivative, exhibits strong tissue protective effects, independent of erythropoietic effects, in a renal ischemia-reperfusion (IR) injury model. Meanwhile, the transforming growth factor-ß (TGF-ß) superfamily member glial cell line-derived neurotrophic factor (GDNF) demonstrated protective effect on podocytes in vitro. Using a rat puromycin aminonucleoside nephropathy (PAN) model, this study observed the renal protective effect of HBSP and investigated its renal protective effect on podocytes and mechanism related to GDNF. METHODS: Rats nephropathy model was induced by injection of 60 mg/kg of PAN via the tail vein. Rats in the PAN + HBSP group were injected intraperitoneally with HBSP (8 nmol/kg) 4 h before the model was induced, followed by intraperitoneal injections of HBSP once every 24 h for 7 consecutive days. The 24-hour urinary protein level was measured once every other day, and blood and renal tissue samples were collected on the 7th day for the examination of renal function, complete blood count, renal pathological changes and the expression levels of GDNF. RESULTS: Compared with the control group, the PAN nephropathy rat model showed a large amount of urinary protein. The pathological manifestations were mainly extensive fusion and disappearance of foot processes, along with vacuolar degeneration of podocytes and their separation from the glomerular basement membrane. GDNF expression was upregulated. Compared with the PAN + vehicle group, the PAN + HBSP group showed decreased urinary protein (p < 0.05). Pathological examination revealed ameliorated glomerular injury and vacuolar degeneration of podocytes. The expression of GDNF in the PAN nephropathy group was increased, when compared with the control group. The greatest expression of GDNF observed in the PAN + HBSP group (p < 0.05). CONCLUSIONS: The expression of GDNF in the kidney of PAN rat model was increased. HBSP reduced urinary protein, ameliorated pathological changes in renal podocytes, increased the expression of GDNF in the PAN rat model. HBSP is likely to exert its protective effects on podocytes through upregulation of GDNF expression.


Subject(s)
Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor , Kidney Diseases , Podocytes , Puromycin Aminonucleoside , Rats, Sprague-Dawley , Animals , Rats , Podocytes/drug effects , Podocytes/pathology , Podocytes/metabolism , Male , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Kidney Diseases/pathology , Kidney/pathology , Kidney/drug effects , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Erythropoietin , Peptide Fragments
8.
J Pathol ; 264(2): 212-227, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39177649

ABSTRACT

WT1 encodes a podocyte transcription factor whose variants can cause an untreatable glomerular disease in early childhood. Although WT1 regulates many podocyte genes, it is poorly understood which of them are initiators in disease and how they subsequently influence other cell-types in the glomerulus. We hypothesised that this could be resolved using single-cell RNA sequencing (scRNA-seq) and ligand-receptor analysis to profile glomerular cell-cell communication during the early stages of disease in mice harbouring an orthologous human mutation in WT1 (Wt1R394W/+). Podocytes were the most dysregulated cell-type in the early stages of Wt1R394W/+ disease, with disrupted angiogenic signalling between podocytes and the endothelium, including the significant downregulation of transcripts for the vascular factors Vegfa and Nrp1. These signalling changes preceded glomerular endothelial cell loss in advancing disease, a feature also observed in biopsy samples from human WT1 glomerulopathies. Addition of conditioned medium from murine Wt1R394W/+ primary podocytes to wild-type glomerular endothelial cells resulted in impaired endothelial looping and reduced vascular complexity. Despite the loss of key angiogenic molecules in Wt1R394W/+ podocytes, the pro-vascular molecule adrenomedullin was upregulated in Wt1R394W/+ podocytes and plasma and its further administration was able to rescue the impaired looping observed when glomerular endothelium was exposed to Wt1R394W/+ podocyte medium. In comparative analyses, adrenomedullin upregulation was part of a common injury signature across multiple murine and human glomerular disease datasets, whilst other gene changes were unique to WT1 disease. Collectively, our study describes a novel role for altered angiogenic signalling in the initiation of WT1 glomerulopathy. We also identify adrenomedullin as a proangiogenic factor, which despite being upregulated in early injury, offers an insufficient protective response due to the wider milieu of dampened vascular signalling that results in endothelial cell loss in later disease. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Kidney Glomerulus , Podocytes , Signal Transduction , Single-Cell Analysis , Transcriptome , WT1 Proteins , Animals , Podocytes/metabolism , Podocytes/pathology , WT1 Proteins/metabolism , WT1 Proteins/genetics , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/blood supply , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Disease Models, Animal , Mutation , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Adrenomedullin/genetics , Adrenomedullin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Communication , Cells, Cultured
9.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39201721

ABSTRACT

Cell deaths maintain the normal function of tissues and organs. In pathological conditions, the abnormal activation or disruption of cell death often leads to pathophysiological effects. Diabetic kidney disease (DKD), a significant microvascular complication of diabetes, is linked to high mortality and morbidity rates, imposing a substantial burden on global healthcare systems and economies. Loss and detachment of podocytes are key pathological changes in the progression of DKD. This review explores the potential mechanisms of apoptosis, necrosis, autophagy, pyroptosis, ferroptosis, cuproptosis, and podoptosis in podocytes, focusing on how different cell death modes contribute to the progression of DKD. It recognizes the limitations of current research and presents the latest basic and clinical research studies targeting podocyte death pathways in DKD. Lastly, it focuses on the future of targeting podocyte cell death to treat DKD, with the intention of inspiring further research and the development of therapeutic strategies.


Subject(s)
Diabetic Nephropathies , Podocytes , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Podocytes/metabolism , Podocytes/pathology , Animals , Apoptosis , Autophagy , Cell Death
10.
Metabolism ; 159: 155982, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089491

ABSTRACT

BACKGROUND: Receptor-interacting protein kinase (RIPK)3 is an essential molecule for necroptosis and its role in kidney fibrosis has been investigated using various kidney injury models. However, the relevance and the underlying mechanisms of RIPK3 to podocyte injury in albuminuric diabetic kidney disease (DKD) remain unclear. Here, we investigated the role of RIPK3 in glomerular injury of DKD. METHODS: We analyzed RIPK3 expression levels in the kidneys of patients with biopsy-proven DKD and animal models of DKD. Additionally, to confirm the clinical significance of circulating RIPK3, RIPK3 was measured by ELISA in plasma obtained from a prospective observational cohort of patients with type 2 diabetes, and estimated glomerular filtration rate (eGFR) and urine albumin-to-creatinine ratio (UACR), which are indicators of renal function, were followed up during the observation period. To investigate the role of RIPK3 in glomerular damage in DKD, we induced a DKD model using a high-fat diet in Ripk3 knockout and wild-type mice. To assess whether mitochondrial dysfunction and albuminuria in DKD take a Ripk3-dependent pathway, we used single-cell RNA sequencing of kidney cortex and immortalized podocytes treated with high glucose or overexpressing RIPK3. RESULTS: RIPK3 expression was increased in podocytes of diabetic glomeruli with increased albuminuria and decreased podocyte numbers. Plasma RIPK3 levels were significantly elevated in albuminuric diabetic patients than in non-diabetic controls (p = 0.002) and non-albuminuric diabetic patients (p = 0.046). The participants in the highest tertile of plasma RIPK3 had a higher incidence of renal progression (hazard ratio [HR] 2.29 [1.05-4.98]) and incident chronic kidney disease (HR 4.08 [1.10-15.13]). Ripk3 knockout improved albuminuria, podocyte loss, and renal ultrastructure in DKD mice. Increased mitochondrial fragmentation, upregulated mitochondrial fission-related proteins such as phosphoglycerate mutase family member 5 (PGAM5) and dynamin-related protein 1 (Drp1), and mitochondrial ROS were decreased in podocytes of Ripk3 knockout DKD mice. In cultured podocytes, RIPK3 inhibition attenuated mitochondrial fission and mitochondrial dysfunction by decreasing p-mixed lineage kinase domain-like protein (MLKL), PGAM5, and p-Drp1 S616 and mitochondrial translocation of Drp1. CONCLUSIONS: The study demonstrates that RIPK3 reflects deterioration of renal function of DKD. In addition, RIPK3 induces diabetic podocytopathy by regulating mitochondrial fission via PGAM5-Drp1 signaling through MLKL. Inhibition of RIPK3 might be a promising therapeutic option for treating DKD.


Subject(s)
Albuminuria , Diabetic Nephropathies , Mitochondria , Podocytes , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , Animals , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/genetics , Albuminuria/genetics , Albuminuria/metabolism , Mice , Podocytes/metabolism , Podocytes/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Male , Dynamins/genetics , Dynamins/metabolism , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Mice, Inbred C57BL , Female , Middle Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism
11.
J Ethnopharmacol ; 335: 118650, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39094755

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Linggui-Zhugan (LGZG) comprises four herbs and is a classic formula in traditional Chinese medicine. There is strong clinical evidence of its pleiotropic effects in the prevention of diabetes and its related complications. Although several classes of drugs are currently available for clinical management of diabetic kidney disease (DKD), tight glycemic and/or hypertension control may not prevent disease progression. This study evaluated the therapeutic effect of the ethnopharmacological agent LGZG on DKD. AIM OF THE STUDY: This study aimed to investigate the effects of LGZG formula with standard quality control on experimental DKD and its related metabolic disorders in animal model. Meanwhile, the present study aimed to investigate regulatory effects of LGZG on renal proteinase 3 (PR3) to reveal mechanisms underlying renoprotective benefits of LGZG. MATERIALS AND METHODS: LGZG decoction was fingerprinted by high-performance liquid chromatography for quality control. An experimental model of DKD was induced in C57 BL/6J mice by a combination of high-fat diet feeding, uninephrectomy, and intraperitoneal injection of streptozocin. The LGZG decoction was administrated by daily oral gavage. RESULTS: Treatment with LGZG formula significantly attenuated DKD-like traits (including severe albuminuria, mesangial matrix expansion, and podocyte loss) and metabolic dysfunction (disordered body composition and dyslipidemia) in mice. RNA sequencing data revealed a close association of LGZG treatment with marked modulation of signaling pathways related to podocyte injury and cell apoptosis. Mechanistically, LGZG suppressed the DKD-triggered increase in renal PR3 and podocyte apoptosis. In-vitro incubation of mouse immortalized podocytes with LGZG-medicated serum attenuated PR3-mediated apoptosis. CONCLUSION: Our data demonstrated that the LGZG formula protected against DKD in mice and was closely associated with its inhibitory effects on PR3-mediated podocyte apoptosis.


Subject(s)
Apoptosis , Diabetic Nephropathies , Drugs, Chinese Herbal , Mice, Inbred C57BL , Podocytes , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/prevention & control , Diabetic Nephropathies/pathology , Podocytes/drug effects , Podocytes/pathology , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Male , Mice , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications
12.
JCI Insight ; 9(18)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115943

ABSTRACT

Organelle stress exacerbates podocyte injury, contributing to perturbed lipid metabolism. Simultaneous organelle stresses can occur in the kidney in the diseased state; therefore, a thorough analysis of organelle communication is crucial for understanding the progression of kidney diseases. Although organelles closely interact with one another at membrane contact sites, limited studies have explored their involvement in kidney homeostasis. The endoplasmic reticulum (ER) protein, PDZ domain-containing 8 (PDZD8), is implicated in multiple-organelle-tethering processes and cellular lipid homeostasis. In this study, we aimed to elucidate the role of organelle communication in podocyte injury using podocyte-specific Pdzd8-knockout mice. Our findings demonstrated that Pdzd8 deletion exacerbated podocyte injury in an accelerated obesity-related kidney disease model. Proteomic analysis of isolated glomeruli revealed that Pdzd8 deletion exacerbated mitochondrial and endosomal dysfunction during podocyte lipotoxicity. Additionally, electron microscopy revealed the accumulation of abnormal, fatty endosomes in Pdzd8-deficient podocytes during obesity-related kidney diseases. Lipidomic analysis indicated that glucosylceramide accumulated in Pdzd8-deficient podocytes, owing to accelerated production and decelerated degradation. Thus, the organelle-tethering factor, PDZD8, plays a crucial role in maintaining mitochondrial and endosomal homeostasis during podocyte lipotoxicity. Collectively, our findings highlight the importance of organelle communication at the 3-way junction among the ER, mitochondria, and endosomes in preserving podocyte homeostasis.


Subject(s)
Endosomes , Homeostasis , Mice, Knockout , Mitochondria , Podocytes , Podocytes/metabolism , Podocytes/pathology , Animals , Mice , Mitochondria/metabolism , Mitochondria/pathology , Endosomes/metabolism , Lipid Metabolism , Obesity/metabolism , Obesity/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/genetics , Endoplasmic Reticulum/metabolism , Male
13.
Cells ; 13(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39120284

ABSTRACT

Mitochondria are crucial for cellular ATP production. They are highly dynamic organelles, whose morphology and function are controlled through mitochondrial fusion and fission. The specific roles of mitochondria in podocytes, the highly specialized cells of the kidney glomerulus, remain less understood. Given the significant structural, functional, and molecular similarities between mammalian podocytes and Drosophila nephrocytes, we employed fly nephrocytes to explore the roles of mitochondria in cellular function. Our study revealed that alterations in the Pink1-Park (mammalian PINK1-PRKN) pathway can disrupt mitochondrial dynamics in Drosophila nephrocytes. This disruption led to either fragmented or enlarged mitochondria, both of which impaired mitochondrial function. The mitochondrial dysfunction subsequently triggered defective intracellular endocytosis, protein aggregation, and cellular damage. These findings underscore the critical roles of mitochondria in nephrocyte functionality.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Endocytosis , Mitochondria , Mitochondrial Dynamics , Podocytes , Animals , Podocytes/metabolism , Podocytes/pathology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Mitochondria/metabolism , Mitochondria/pathology , Drosophila melanogaster/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases
15.
J Transl Med ; 22(1): 762, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143486

ABSTRACT

BACKGROUND: Personalized disease models are crucial for evaluating how diseased cells respond to treatments, especially in case of innovative biological therapeutics. Extracellular vesicles (EVs), nanosized vesicles released by cells for intercellular communication, have gained therapeutic interest due to their ability to reprogram target cells. We here utilized urinary podocytes obtained from children affected by steroid-resistant nephrotic syndrome with characterized genetic mutations as a model to test the therapeutic potential of EVs derived from kidney progenitor cells (nKPCs). METHODS: EVs were isolated from nKPCs derived from the urine of a preterm neonate. Three lines of urinary podocytes obtained from nephrotic patients' urine and a line of Alport syndrome patient podocytes were characterized and used to assess albumin permeability in response to nKPC-EVs or various drugs. RNA sequencing was conducted to identify commonly modulated pathways after nKPC-EV treatment. siRNA transfection was used to demonstrate the involvement of SUMO1 and SENP2 in the modulation of permeability. RESULTS: Treatment with the nKPC-EVs significantly reduced permeability across all the steroid-resistant patients-derived and Alport syndrome-derived podocytes. At variance, podocytes appeared unresponsive to standard pharmacological treatments, with the exception of one line, in alignment with the patient's clinical response at 48 months. By RNA sequencing, only two genes were commonly upregulated in nKPC-EV-treated genetically altered podocytes: small ubiquitin-related modifier 1 (SUMO1) and Sentrin-specific protease 2 (SENP2). SUMO1 and SENP2 downregulation increased podocyte permeability confirming the role of the SUMOylation pathway. CONCLUSIONS: nKPCs emerge as a promising non-invasive source of EVs with potential therapeutic effects on podocytes with genetic dysfunction, through modulation of SUMOylation, an important pathway for the stability of podocyte slit diaphragm proteins. Our findings also suggest the feasibility of developing a non-invasive in vitro model for screening regenerative compounds on patient-derived podocytes.


Subject(s)
Extracellular Vesicles , Nephrotic Syndrome , Podocytes , Podocytes/metabolism , Podocytes/drug effects , Podocytes/pathology , Humans , Nephrotic Syndrome/pathology , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/metabolism , Extracellular Vesicles/metabolism , Drug Evaluation, Preclinical , Models, Biological , Stem Cells/metabolism , Steroids/pharmacology , Kidney/pathology , Kidney/metabolism , Drug Resistance , Infant, Newborn , Male
16.
J Clin Invest ; 134(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087471

ABSTRACT

Glomerular visceral epithelial cells (i.e., podocytes) are an essential component of the tripartite glomerular filtration barrier. Healthy podocytes are terminally differentiated cells with limited replicative capacity; however, inappropriate cell cycle reentry can be induced in podocytes by various injurious stimuli. In this issue of the JCI, Yamaguchi et al. report on a somatic mosaic gain-of-function mutation in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic α subunit (p110α, encoded by PIK3CA). The study reveals that activating mutations of p110α can drive podocyte proliferation in PIK3CA-related overgrowth syndrome (PROS). They also showed that selective, small-molecule inhibitors of p110 may be useful for the treatment of proliferative glomerulonephritis.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Phosphatidylinositol 3-Kinases , Podocytes , Humans , Podocytes/pathology , Podocytes/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Cell Proliferation , Mice , Mutation , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/pathology , Glomerulonephritis, Membranoproliferative/immunology
17.
Kidney Int ; 106(3): 359-361, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174196

ABSTRACT

Alport syndrome is a hereditary kidney disease caused by collagen IV mutations that interfere with the formation and deposition of the α3α4α5 protomer into the glomerular basement membrane. In this issue, Yu et al. show that the chemical chaperone tauroursodeoxycholic acid prevented kidney structural changes and function decline in mice with a pathogenic missense Col4a3 mutation by increasing mutant α3α4α5 protomer glomerular basement membrane deposition and preventing podocyte apoptosis induced by endoplasmic reticulum stress.


Subject(s)
Autoantigens , Collagen Type IV , Glomerular Basement Membrane , Nephritis, Hereditary , Taurochenodeoxycholic Acid , Nephritis, Hereditary/genetics , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/pathology , Nephritis, Hereditary/metabolism , Animals , Collagen Type IV/genetics , Collagen Type IV/metabolism , Taurochenodeoxycholic Acid/pharmacology , Taurochenodeoxycholic Acid/therapeutic use , Mice , Glomerular Basement Membrane/pathology , Glomerular Basement Membrane/drug effects , Humans , Autoantigens/genetics , Autoantigens/metabolism , Endoplasmic Reticulum Stress/drug effects , Apoptosis/drug effects , Disease Models, Animal , Podocytes/drug effects , Podocytes/pathology , Podocytes/metabolism , Mutation, Missense , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
18.
Clin Exp Pharmacol Physiol ; 51(9): e13909, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39038854

ABSTRACT

Diabetic kidney disease (DKD) is a complication of diabetic mellitus. New treatments need to be developed. This study aimed to investigate the effects of quercetin-4'-O-ß-D-glucopyranoside (QODG) on podocyte injury. Podocytes were cultured in high glucose (HG) medium, treated with QODG, and overexpressing or knocking down SIRT5. Oxidative stress indicators were assessed using corresponding kits. Pyroptosis was detected by flow cytometry and western blot analysis. Succinylation modification was detected using immunoprecipitation (IP) and western blot analysis. The interaction between NEK7 and NLRP3 was determined by co-IP. The results indicated that QODG inhibited oxidative stress and pyroptosis of podocytes induced by HG. Besides, QODG suppressed succinylation levels in HG-induced podocytes, with the upregulation of SIRT5. Knockdown of SIRT5 reversed the effects of QODG on oxidative stress and pyroptosis. Moreover, SIRT5 inhibited the succinylation of NEK7 and the interaction between NLRP3 and NEK7. In conclusion, QODG upregulates SIRT5 to inhibit the succinylation modification of NEK7, impedes the interaction between NEK7 and NLRP3, and then inhibits the pyroptosis and oxidative stress injury of podocytes under HG conditions. The findings suggested that QODG has the potential to treat DKD and explore a novel underlying mechanism of QODG function.


Subject(s)
NIMA-Related Kinases , Podocytes , Sirtuins , Podocytes/drug effects , Podocytes/metabolism , Podocytes/pathology , NIMA-Related Kinases/metabolism , Sirtuins/metabolism , Sirtuins/genetics , Animals , Mice , Oxidative Stress/drug effects , Pyroptosis/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Glucosides/pharmacology , Cell Line
19.
Kidney Blood Press Res ; 49(1): 605-618, 2024.
Article in English | MEDLINE | ID: mdl-38952124

ABSTRACT

INTRODUCTION: The calcineurin inhibitor cyclosporine A (CsA) has been shown to effectively reduce proteinuria. However, its precise mechanism is still not fully understood. Our previous study showed that CsA reduced proteinuria by directly stabilizing the foot process (FP) cytoskeletal structure via cofilin-1, suggesting that synaptopodin, a podocyte-specific actin protein, is not the sole target of CsA in podocytes. METHODS: In this study, we established an adriamycin (ADR)-induced nephropathy rat model and a cultured podocyte injury model. We employed Western blotting and immunofluorescence techniques to assess the expression and distribution of transgelin, Krüppel-like factor-4 (KLF-4), nephrin, and synaptopodin. RESULTS: We observed a significant increase in proteinuria levels accompanied by loss of normal FP structure in the ADR-induced nephropathy rat model. The levels of the actin cross-linking protein transgelin were increased significantly, while those of the podocyte-specific molecules nephrin and synaptopodin were decreased in vivo. Treatment with CsA effectively reduced proteinuria while restoring FP effacement stability in ADR-induced nephropathy models and restoring the expression of transgelin, nephrin, and synaptopodin both in vivo and in vitro. Furthermore, CsA treatment dose-dependently decreased transgelin levels while significantly increasing KLF-4 expression in injured podocytes. In addition, CsA failed to downregulate transgelin when KLF-4 was specifically knocked down. CONCLUSION: Our findings suggest that CsA protects against podocyte injury by downregulating abnormally high levels of transgelin via upregulation of KLF-4 expression.


Subject(s)
Cyclosporine , Doxorubicin , Kruppel-Like Factor 4 , Microfilament Proteins , Muscle Proteins , Podocytes , Podocytes/drug effects , Podocytes/pathology , Podocytes/metabolism , Animals , Microfilament Proteins/metabolism , Rats , Cyclosporine/pharmacology , Kruppel-Like Factor 4/metabolism , Muscle Proteins/metabolism , Muscle Proteins/biosynthesis , Male , Membrane Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Rats, Sprague-Dawley , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Kidney Diseases/metabolism , Kidney Diseases/pathology , Proteinuria
20.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995008

ABSTRACT

Accumulating evidence indicates that inflammatory and immunologic processes play a significant role in the development and progression of glomerular diseases. Podocytes, the terminally differentiated epithelial cells, are crucial for maintaining the integrity of the glomerular filtration barrier. Once injured, podocytes cannot regenerate, leading to progressive proteinuric glomerular diseases. However, emerging evidence suggests that podocytes not only maintain the glomerular filtration barrier and are important targets of immune responses but also exhibit many features of immune-like cells, where they are involved in the modulation of the activity of innate and adaptive immunity. This dual role of podocytes may lead to the discovery and development of new therapeutic targets for treating glomerular diseases. This review aims to provide an overview of the innate immunity mechanisms involved in podocyte injury and the progression of proteinuric glomerular diseases.


Subject(s)
Immunity, Innate , Podocytes , Podocytes/immunology , Podocytes/pathology , Humans , Animals , Kidney Diseases/immunology , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Kidney Glomerulus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL