Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.261
Filtrer
1.
Nat Commun ; 15(1): 8038, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39271683

RÉSUMÉ

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.


Sujet(s)
Néphropathies diabétiques , Tubules contournés proximaux , Facteur-6 de type krüppel , Podocytes , Transduction du signal , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/génétique , Podocytes/métabolisme , Podocytes/anatomopathologie , Animaux , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Mâle , Humains , Souris , Facteur-6 de type krüppel/métabolisme , Facteur-6 de type krüppel/génétique , Calcium-Calmodulin-Dependent Protein Kinase Type 1/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 1/génétique , Souris de lignée C57BL , Modèles animaux de maladie humaine
2.
Int J Biol Sci ; 20(12): 4654-4673, 2024.
Article de Anglais | MEDLINE | ID: mdl-39309435

RÉSUMÉ

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.


Sujet(s)
Néphropathies diabétiques , microARN , Mitochondries , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Podocytes , Podocytes/métabolisme , Podocytes/anatomopathologie , microARN/métabolisme , microARN/génétique , Animaux , Souris , Mitochondries/métabolisme , Humains , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Mâle , Souris de lignée C57BL , Femelle
3.
Cells ; 13(17)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39273018

RÉSUMÉ

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.


Sujet(s)
Autophagie , Doxorubicine , Souris knockout , Récepteur PPAR alpha , Podocytes , Animaux , Podocytes/métabolisme , Podocytes/anatomopathologie , Podocytes/effets des médicaments et des substances chimiques , Doxorubicine/effets indésirables , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Souris , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire , Maladies du rein/induit chimiquement , Maladies du rein/anatomopathologie , Maladies du rein/métabolisme , Maladies du rein/génétique , Apoptose/effets des médicaments et des substances chimiques , Fénofibrate/pharmacologie , Mâle
4.
Mol Biol Rep ; 51(1): 1003, 2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-39305364

RÉSUMÉ

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.


Sujet(s)
AMP-Activated Protein Kinases , Autophagie , Néphropathies diabétiques , Glucose , Protéines membranaires , Podocytes , Transduction du signal , Triterpènes , Podocytes/effets des médicaments et des substances chimiques , Podocytes/métabolisme , Podocytes/anatomopathologie , AMP-Activated Protein Kinases/métabolisme , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Autophagie/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Triterpènes/pharmacologie , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
5.
Biol Sex Differ ; 15(1): 72, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39278930

RÉSUMÉ

BACKGROUND: Sex differences exist in the prevalence and progression of major glomerular diseases. Podocytes are the essential cell-type in the kidney which maintain the physiological blood-urine barrier, and pathological changes in podocyte homeostasis are critical accelerators of impairment of kidney function. However, sex-specific molecular signatures of podocytes under physiological and stress conditions remain unknown. This work aimed at identifying sexual dimorphic molecular signatures of podocytes under physiological condition and pharmacologically challenged homeostasis with mechanistic target of rapamycin (mTOR) inhibition. mTOR is a crucial regulator involved in a variety of physiological and pathological stress responses in the kidney and inhibition of this pathway may therefore serve as a general stress challenger to get fundamental insights into sex differences in podocytes. METHODS: The genomic ROSAmT/mG-NPHS2 Cre mouse model was used which allows obtaining highly pure podocyte fractions for cell-specific molecular analyses, and vehicle or pharmacologic treatment with the mTOR inhibitor rapamycin was performed for 3 weeks. Subsequently, deep RNA sequencing and proteomics were performed of the isolated podocytes to identify intrinsic sex differences. Studies were supplemented with metabolomics from kidney cortex tissues. RESULTS: Although kidney function and morphology remained normal in all experimental groups, RNA sequencing, proteomics and metabolomics revealed strong intrinsic sex differences in the expression levels of mitochondrial, translation and structural transcripts, protein abundances and regulation of metabolic pathways. Interestingly, rapamycin abolished prominent sex-specific clustering of podocyte gene expression and induced major changes only in male transcriptome. Several sex-biased transcription factors could be identified as possible upstream regulators of these sexually dimorphic responses. Concordant to transcriptomics, metabolomic changes were more prominent in males. Remarkably, high number of previously reported kidney disease genes showed intrinsic sexual dimorphism and/or different response patterns towards mTOR inhibition. CONCLUSIONS: Our results highlight remarkable intrinsic sex-differences and sex-specific response patterns towards pharmacological challenged podocyte homeostasis which might fundamentally contribute to sex differences in kidney disease susceptibilities and progression. This work provides rationale and an in-depth database for novel targets to be tested in specific kidney disease models to advance with sex-specific treatment strategies.


The global burden of chronic kidney diseases is rapidly increasing and is projected to become the fifth most common cause of years of life lost worldwide by 2040. Sexual dimorphism in kidney diseases and transplantation is well known, yet sex-specific therapeutic strategies are still missing. One reason is the lack of knowledge due to the lack of inclusion of sex as a biological variable in study designs. This work aimed at identification of molecular signatures of male and female podocytes, gate-keepers of the glomerular filtration barrier. Like cardiomyocytes, podocytes are terminally differentiated cells which are highly susceptible towards pathological challenges. Podocytes are the decisive cell-type of the kidney to maintain the physiological blood-urine barrier, and disturbances of their homeostasis critically accelerate kidney function impairment. By help of a genomic mouse model, highly purified podocytes were obtained from male and female mice with and without pharmacological challenge of the mechanistic target of rapamycin (mTOR) signaling pathway which is known to be deregulated in major kidney diseases. Deep RNA sequencing, proteomics and metabolomics revealed strong intrinsic sex differences in the expression levels of mitochondrial, translation and structural transcripts, protein abundances and regulation of metabolic pathways which might fundamentally contribute to sex differences in kidney disease susceptibilities and progression. Remarkably, high number of previously reported kidney disease genes showed so far unknown intrinsic sexual dimorphism and/or different response patterns towards mTOR inhibition. Our work provides an in-depth database for novel targets to be tested in kidney disease models to advance with sex-specific treatment strategies.


Sujet(s)
Homéostasie , Podocytes , Caractères sexuels , Sirolimus , Animaux , Podocytes/effets des médicaments et des substances chimiques , Podocytes/métabolisme , Mâle , Femelle , Sirolimus/pharmacologie , Homéostasie/effets des médicaments et des substances chimiques , Souris , Sérine-thréonine kinases TOR/métabolisme , Transcriptome , Inhibiteurs de mTOR/pharmacologie
6.
Ren Fail ; 46(2): 2398712, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39248407

RÉSUMÉ

As an important component of the glomerular filtration membrane, the state of the podocytes is closely related to kidney function, they are also key cells involved in aging and play a central role in the damage caused by renal aging. Therefore, understanding the aging process of podocytes will allow us to understand their susceptibility to injury and identify targeted protective mechanisms. In fact, the process of physiological aging itself can induce podocyte senescence. Pathological stresses, such as oxidative stress, mitochondrial damage, secretion of senescence-associated secretory phenotype, reduced autophagy, oncogene activation, altered transcription factors, DNA damage response, and other factors, play a crucial role in inducing premature senescence and accelerating aging. Senescence-associated-ß-galactosidase (SA-ß-gal) is a marker of aging, and ß-hydroxybutyric acid treatment can reduce SA-ß-gal activity to alleviate cellular senescence and damage. In addition, CCAAT/enhancer-binding protein-α, transforming growth factor-ß signaling, glycogen synthase kinase-3ß, cycle-dependent kinase, programmed cell death protein 1, and plasminogen activator inhibitor-1 are closely related to aging. The absence or elevation of these factors can affect aging through different mechanisms. Podocyte injury is not an independent process, and injured podocytes interact with the surrounding epithelial cells or other kidney cells to mediate the injury or loss of podocytes. In this review, we discuss the manifestations, molecular mechanisms, biomarkers, and therapeutic drugs for podocyte senescence. We included elamipretide, lithium, calorie restriction, rapamycin; and emerging treatment strategies, such as gene and immune therapies. More importantly, we summarize how podocyte interact with other kidney cells.


Sujet(s)
Vieillissement de la cellule , Podocytes , Podocytes/métabolisme , Humains , Stress oxydatif , Animaux , Vieillissement/métabolisme , Transduction du signal , Autophagie
7.
J Vis Exp ; (210)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39248530

RÉSUMÉ

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.


Sujet(s)
Apoptose , Médicaments issus de plantes chinoises , Glomérulonéphrite extra-membraneuse , Phosphatidylinositol 3-kinases , Podocytes , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Glomérulonéphrite extra-membraneuse/anatomopathologie , Glomérulonéphrite extra-membraneuse/traitement médicamenteux , Glomérulonéphrite extra-membraneuse/métabolisme , Podocytes/effets des médicaments et des substances chimiques , Podocytes/métabolisme , Podocytes/anatomopathologie , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Mâle , Rat Sprague-Dawley , Morpholines/pharmacologie , Morpholines/usage thérapeutique , 4H-1-Benzopyran-4-ones/pharmacologie , Modèles animaux de maladie humaine
8.
Endocrinology ; 165(10)2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39133576

RÉSUMÉ

The phosphorylation of the transmembrane protein nephrin has been shown to play an important role in signaling in kidney podocytes, and it has now been shown to also play a key role in regulating pancreatic ß-cell function. Williamson et al have recently shown that the loss of nephrin tyrosine phosphorylation on its 3 cytoplasmic YDxV motifs can enhance insulin release in aged female mice. These studies suggest that blocking nephrin phosphorylation may be an effective treatment option for improving ß-cell function.


Sujet(s)
Cellules à insuline , Protéines membranaires , Animaux , Cellules à insuline/métabolisme , Cellules à insuline/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Souris , Femelle , Humains , Insuline/métabolisme , Podocytes/métabolisme , Podocytes/effets des médicaments et des substances chimiques
9.
JCI Insight ; 9(18)2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39115943

RÉSUMÉ

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.


Sujet(s)
Endosomes , Homéostasie , Souris knockout , Mitochondries , Podocytes , Podocytes/métabolisme , Podocytes/anatomopathologie , Animaux , Souris , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Endosomes/métabolisme , Métabolisme lipidique , Obésité/métabolisme , Obésité/anatomopathologie , Maladies du rein/métabolisme , Maladies du rein/anatomopathologie , Maladies du rein/génétique , Réticulum endoplasmique/métabolisme , Mâle
10.
Physiol Genomics ; 56(10): 672-690, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39101921

RÉSUMÉ

Atrial and brain natriuretic peptides (ANP and BNP) bind to guanylyl cyclase A/natriuretic peptide receptor A (GC-A/NPRA), stimulating natriuresis and diuresis and reducing blood pressure (BP), but the role of ANP/NPRA signaling in podocytes (highly specialized epithelial cells covering the outer surfaces of renal glomerular capillaries) remains unclear. This study aimed to determine the effect of conditional deletion of podocyte-specific Npr1 (encoding NPRA) gene knockout (KO) in male and female mice. Tamoxifen-treated wild-type control (PD Npr1 f/f; WT), heterozygous (PD-Cre-Npr1 f/+; HT), and KO (PD-Cre-Npr1 f/-) mice were fed a normal-, low-, or high-salt diet for 4 wk. Podocytes isolated from HT and KO male and female mice showed complete absence of Npr1 mRNA and NPRA protein compared with WT mice. BP, plasma creatinine, plasma sodium, urinary protein, and albumin/creatinine ratio were significantly increased, whereas plasma total protein, albumin, creatinine clearance, and urinary sodium levels were significantly reduced in the HT and KO male and female mice compared with WT mice. These changes were significantly greater in males than in females. On a normal-salt diet, glomerular filtration rate was significantly decreased in PD Npr1 HT and KO male and female mice compared with WT mice. Immunofluorescence of podocin and synaptopodin was also significantly reduced in HT and KO mice compared with WT mice. These observations suggest that in podocytes, ANP/NPRA signaling may be crucial in the maintenance and regulation of glomerular filtration and BP and serve as a biomarker of renal function in a sex-dependent manner.NEW & NOTEWORTHY Our results demonstrate that the podocyte-specific deletion of Npr1 showed increased blood pressure (BP) and altered biomarkers of renal functions, with greater magnitudes in animals fed a high-salt diet in a sex-dependent manner. The results suggest a direct and sex-dependent effect of Npr1 ablation in podocytes on the regulation of BP and renal function and reveal that podocytes may be considered an important target for the ANP-BNP/NPRA/cGMP signaling cascade.


Sujet(s)
Pression sanguine , Homéostasie , Rein , Souris knockout , Podocytes , Récepteur facteur natriurétique auriculaire , Animaux , Femelle , Récepteur facteur natriurétique auriculaire/génétique , Récepteur facteur natriurétique auriculaire/métabolisme , Mâle , Podocytes/métabolisme , Souris , Rein/métabolisme , Caractères sexuels , Facteurs sexuels , Transduction du signal
11.
Elife ; 122024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39207915

RÉSUMÉ

HIV disease remains prevalent in the USA and chronic kidney disease remains a major cause of morbidity in HIV-1-positive patients. Host double-stranded RNA (dsRNA)-activated protein kinase (PKR) is a sensor for viral dsRNA, including HIV-1. We show that PKR inhibition by compound C16 ameliorates the HIV-associated nephropathy (HIVAN) kidney phenotype in the Tg26 transgenic mouse model, with reversal of mitochondrial dysfunction. Combined analysis of single-nucleus RNA-seq and bulk RNA-seq data revealed that oxidative phosphorylation was one of the most downregulated pathways and identified signal transducer and activator of transcription (STAT3) as a potential mediating factor. We identified in Tg26 mice a novel proximal tubular cell cluster enriched in mitochondrial transcripts. Podocytes showed high levels of HIV-1 gene expression and dysregulation of cytoskeleton-related genes, and these cells dedifferentiated. In injured proximal tubules, cell-cell interaction analysis indicated activation of the pro-fibrogenic PKR-STAT3-platelet-derived growth factor (PDGF)-D pathway. These findings suggest that PKR inhibition and mitochondrial rescue are potential novel therapeutic approaches for HIVAN.


Sujet(s)
Néphropathie associée au SIDA , Souris transgéniques , Mitochondries , eIF-2 Kinase , Animaux , Humains , Souris , Néphropathie associée au SIDA/génétique , Néphropathie associée au SIDA/métabolisme , Néphropathie associée au SIDA/anatomopathologie , Modèles animaux de maladie humaine , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Mitochondries/métabolisme , Podocytes/métabolisme , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique
12.
Cells ; 13(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39120284

RÉSUMÉ

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.


Sujet(s)
Protéines de Drosophila , Drosophila melanogaster , Endocytose , Mitochondries , Dynamique mitochondriale , Podocytes , Animaux , Podocytes/métabolisme , Podocytes/anatomopathologie , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Drosophila melanogaster/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Ubiquitin-protein ligases
13.
Ren Fail ; 46(2): 2394637, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39189638

RÉSUMÉ

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.


Sujet(s)
Modèles animaux de maladie humaine , Facteur neurotrophique dérivé des cellules gliales , Maladies du rein , Podocytes , Puromycine aminonucléoside , Rat Sprague-Dawley , Animaux , Rats , Podocytes/effets des médicaments et des substances chimiques , Podocytes/anatomopathologie , Podocytes/métabolisme , Mâle , Facteur neurotrophique dérivé des cellules gliales/métabolisme , Maladies du rein/induit chimiquement , Maladies du rein/prévention et contrôle , Maladies du rein/anatomopathologie , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/métabolisme , Érythropoïétine , Fragments peptidiques
14.
J Transl Med ; 22(1): 762, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143486

RÉSUMÉ

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.


Sujet(s)
Vésicules extracellulaires , Syndrome néphrotique , Podocytes , Podocytes/métabolisme , Podocytes/effets des médicaments et des substances chimiques , Podocytes/anatomopathologie , Humains , Syndrome néphrotique/anatomopathologie , Syndrome néphrotique/traitement médicamenteux , Syndrome néphrotique/métabolisme , Vésicules extracellulaires/métabolisme , Évaluation préclinique de médicament , Modèles biologiques , Cellules souches/métabolisme , Stéroïdes/pharmacologie , Rein/anatomopathologie , Rein/métabolisme , Résistance aux substances , Nouveau-né , Mâle
15.
Kidney Int ; 106(3): 359-361, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39174196

RÉSUMÉ

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.


Sujet(s)
Autoantigènes , Collagène de type IV , Membrane basale glomérulaire , Néphropathie familiale avec surdité , Acide taurochénodésoxycholique , Néphropathie familiale avec surdité/génétique , Néphropathie familiale avec surdité/traitement médicamenteux , Néphropathie familiale avec surdité/anatomopathologie , Néphropathie familiale avec surdité/métabolisme , Animaux , Collagène de type IV/génétique , Collagène de type IV/métabolisme , Acide taurochénodésoxycholique/pharmacologie , Acide taurochénodésoxycholique/usage thérapeutique , Souris , Membrane basale glomérulaire/anatomopathologie , Membrane basale glomérulaire/effets des médicaments et des substances chimiques , Humains , Autoantigènes/génétique , Autoantigènes/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Podocytes/effets des médicaments et des substances chimiques , Podocytes/anatomopathologie , Podocytes/métabolisme , Mutation faux-sens , Chaperons moléculaires/génétique , Chaperons moléculaires/métabolisme
16.
Front Immunol ; 15: 1441017, 2024.
Article de Anglais | MEDLINE | ID: mdl-39185424

RÉSUMÉ

The most characteristic feature of membranous nephropathy (MN) is the presence of subepithelial electron dense deposits and the consequential thickening of the glomerular basement membrane. There have been great advances in the understanding of the destiny of immune complexes in MN by the benefit of experimental models represented by Heymann nephritis. Subepithelial immune complexes are formed in situ by autoantibodies targeting native autoantigens or exogenous planted antigens such as the phospholipase A2 receptor (PLA2R) and cationic BSA respectively. The nascent immune complexes would not be pathogenic until they develop into immune deposits. Podocytes are the major source of autoantigens in idiopathic membranous nephropathy. They also participate in the modulation and removal of the immune complexes to a large extent. The balance between deposition and clearance is regulated by a wide range of factors such as the composition and physicochemical properties of the immune complexes and the complement system. Complement components such as C3 and C1q have been reported to be precipitated with the deposits whereas a complement regulatory protein CR1 expressed by podocytes is involved in the phagocytosis of immune complexes by podocytes. Podocytes regulate the dynamic change of immune complexes which is disturbed in membranous nephropathy. To elucidate the precise fate of the immune complexes is essential for developing more rational and novel therapies for membranous nephropathy.


Sujet(s)
Complexe antigène-anticorps , Glomérulonéphrite extra-membraneuse , Podocytes , Glomérulonéphrite extra-membraneuse/immunologie , Humains , Complexe antigène-anticorps/immunologie , Complexe antigène-anticorps/métabolisme , Podocytes/immunologie , Podocytes/métabolisme , Animaux , Autoanticorps/immunologie , Autoantigènes/immunologie , Récepteurs à la phospholipase A2/immunologie
17.
Cells ; 13(16)2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39195240

RÉSUMÉ

In nephrotic syndrome, the podocyte filtration structures are damaged in a process called foot process effacement. This is mediated by the actin cytoskeleton; however, which actins are involved and how they interact with other filtration components, like the basement membrane, remains poorly understood. Here, we used the well-established Drosophila pericardial nephrocyte-the equivalent of podocytes in flies-knockdown models (RNAi) to study the interplay of the actin cytoskeleton (Act5C, Act57B, Act42A, and Act87E), alpha- and beta-integrin (basement membrane), and the slit diaphragm (Sns and Pyd). Knockdown of an actin gene led to variations of formation of actin stress fibers, the internalization of Sns, and a disrupted slit diaphragm cortical pattern. Notably, deficiency of Act5C, which resulted in complete absence of nephrocytes, could be partially mitigated by overexpressing Act42A or Act87E, suggesting at least partial functional redundancy. Integrin localized near the actin cytoskeleton as well as slit diaphragm components, but when the nephrocyte cytoskeleton or slit diaphragm was disrupted, this switched to colocalization, both at the surface and internalized in aggregates. Altogether, the data show that the interdependence of the slit diaphragm, actin cytoskeleton, and integrins is key to the structure and function of the Drosophila nephrocyte.


Sujet(s)
Cytosquelette d'actine , Protéines de Drosophila , Intégrines , Podocytes , Animaux , Cytosquelette d'actine/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Intégrines/métabolisme , Podocytes/métabolisme , Drosophila melanogaster/métabolisme , Drosophila/métabolisme , Actines/métabolisme , Immunoglobulines
18.
J Pathol ; 264(2): 212-227, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39177649

RÉSUMÉ

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.


Sujet(s)
Glomérule rénal , Podocytes , Transduction du signal , Analyse sur cellule unique , Transcriptome , Protéines WT1 , Animaux , Podocytes/métabolisme , Podocytes/anatomopathologie , Protéines WT1/métabolisme , Protéines WT1/génétique , Humains , Glomérule rénal/métabolisme , Glomérule rénal/anatomopathologie , Glomérule rénal/vascularisation , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Souris , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Modèles animaux de maladie humaine , Mutation , Maladies du rein/génétique , Maladies du rein/métabolisme , Maladies du rein/anatomopathologie , Adrénomédulline/génétique , Adrénomédulline/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Communication cellulaire , Cellules cultivées
19.
Metabolism ; 159: 155982, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39089491

RÉSUMÉ

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.


Sujet(s)
Albuminurie , Néphropathies diabétiques , Mitochondries , Podocytes , Receptor-Interacting Protein Serine-Threonine Kinases , Transduction du signal , Animaux , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/génétique , Albuminurie/génétique , Albuminurie/métabolisme , Souris , Podocytes/métabolisme , Podocytes/anatomopathologie , Humains , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Mâle , Dynamines/génétique , Dynamines/métabolisme , Souris knockout , Phosphoprotein Phosphatases/génétique , Phosphoprotein Phosphatases/métabolisme , Souris de lignée C57BL , Femelle , Adulte d'âge moyen , Diabète de type 2/complications , Diabète de type 2/métabolisme
20.
J Clin Invest ; 134(15)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39087471

RÉSUMÉ

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.


Sujet(s)
Phosphatidylinositol 3-kinases de classe I , Phosphatidylinositol 3-kinases , Podocytes , Humains , Podocytes/anatomopathologie , Podocytes/métabolisme , Animaux , Phosphatidylinositol 3-kinases de classe I/génétique , Phosphatidylinositol 3-kinases de classe I/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Prolifération cellulaire , Souris , Mutation , Glomérulonéphrite/génétique , Glomérulonéphrite/anatomopathologie , Glomérulonéphrite membranoproliférative/génétique , Glomérulonéphrite membranoproliférative/anatomopathologie , Glomérulonéphrite membranoproliférative/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE