Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.490
Filtrer
2.
Cell Commun Signal ; 22(1): 351, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970061

RÉSUMÉ

BACKGROUND: Accompanied by activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, aberrant connexin 43 (Cx43) hemichannel-mediated ATP release is situated upstream of inflammasome assembly and inflammation and contributes to multiple secondary complications of diabetes and associated cardiometabolic comorbidities. Evidence suggests there may be a link between Cx43 hemichannel activity and inflammation in the diabetic kidney. The consequences of blocking tubular Cx43 hemichannel-mediated ATP release in priming/activation of the NLRP3 inflammasome in a model of diabetic kidney disease (DKD) was investigated. We examined downstream markers of inflammation and the proinflammatory and chemoattractant role of the tubular secretome on macrophage recruitment and activation. METHODS: Analysis of human transcriptomic data from the Nephroseq repository correlated gene expression to renal function in DKD. Primary human renal proximal tubule epithelial cells (RPTECs) and monocyte-derived macrophages (MDMs) were cultured in high glucose and inflammatory cytokines as a model of DKD to assess Cx43 hemichannel activity, NLRP3 inflammasome activation and epithelial-to-macrophage paracrine-mediated crosstalk. Tonabersat assessed a role for Cx43 hemichannels. RESULTS: Transcriptomic analysis from renal biopsies of patients with DKD showed that increased Cx43 and NLRP3 expression correlated with declining glomerular filtration rate (GFR) and increased proteinuria. In vitro, Tonabersat blocked glucose/cytokine-dependant increases in Cx43 hemichannel-mediated ATP release and reduced expression of inflammatory markers and NLRP3 inflammasome activation in RPTECs. We observed a reciprocal relationship in which NLRP3 activity exacerbated increased Cx43 expression and hemichannel-mediated ATP release, events driven by nuclear factor kappa-B (NFκB)-mediated priming and Cx43 hemichannel opening, changes blocked by Tonabersat. Conditioned media (CM) from RPTECs treated with high glucose/cytokines increased expression of inflammatory markers in MDMs, an effect reduced when macrophages were pre-treated with Tonabersat. Co-culture using conditioned media from Tonabersat-treated RPTECs dampened macrophage inflammatory marker expression and reduced macrophage migration. CONCLUSION: Using a model of DKD, we report for the first time that high glucose and inflammatory cytokines trigger aberrant Cx43 hemichannel activity, events that instigate NLRP3-induced inflammation in RPTECs and epithelial-to-macrophage crosstalk. Recapitulating observations previously reported in diabetic retinopathy, these data suggest that Cx43 hemichannel blockers (i.e., Tonabersat) may dampen multi-system damage observed in secondary complications of diabetes.


Sujet(s)
Néphropathies diabétiques , Inflammasomes , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Humains , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Inflammasomes/métabolisme , Connexine 43/métabolisme , Connexine 43/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Adénosine triphosphate/métabolisme , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie
3.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38928090

RÉSUMÉ

Nicotinamide adenine dinucleotide (NAD) is involved in renal physiology and is synthesized by nicotinamide mononucleotide adenylyltransferase (NMNAT). NMNAT exists as three isoforms, namely, NMNAT1, NMNAT2, and NMNAT3, encoded by Nmnat1, Nmnat2, and Nmnat3, respectively. In diabetic nephropathy (DN), NAD levels decrease, aggravating renal fibrosis. Conversely, sodium-glucose cotransporter-2 inhibitors increase NAD levels, mitigating renal fibrosis. In this regard, renal NAD synthesis has recently gained attention. However, the renal role of Nmnat in DN remains uncertain. Therefore, we investigated the role of Nmnat by establishing genetically engineered mice. Among the three isoforms, NMNAT1 levels were markedly reduced in the proximal tubules (PTs) of db/db mice. We examined the phenotypic changes in PT-specific Nmnat1 conditional knockout (CKO) mice. In CKO mice, Nmnat1 expression in PTs was downregulated when the tubules exhibited albuminuria, peritubular type IV collagen deposition, and mitochondrial ribosome (mitoribosome) excess. In CKO mice, Nmnat1 deficiency-induced mitoribosome excess hindered mitoribosomal translation of mitochondrial inner membrane-associated oxidative phosphorylation complex I (CI), CIII, CIV, and CV proteins and mitoribosomal dysfunction. Furthermore, the expression of hypermethylated in cancer 1, a transcription repressor, was downregulated in CKO mice, causing mitoribosome excess. Nmnat1 overexpression preserved mitoribosomal function, suggesting its protective role in DN.


Sujet(s)
Néphropathies diabétiques , Souris knockout , Nicotinamide nucleotide adenylyltransferase , Animaux , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/génétique , Néphropathies diabétiques/anatomopathologie , Souris , Nicotinamide nucleotide adenylyltransferase/métabolisme , Nicotinamide nucleotide adenylyltransferase/génétique , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Mâle , Mitochondries/métabolisme , Souris de lignée C57BL
4.
Sci Rep ; 14(1): 14552, 2024 06 24.
Article de Anglais | MEDLINE | ID: mdl-38914593

RÉSUMÉ

We have reported that an environmental pollutant, cadmium, promotes cell death in the human renal tubular cells (RTCs) through hyperactivation of a serine/threonine kinase Akt. However, the molecular mechanisms downstream of Akt in this process have not been elucidated. Cadmium has a potential to accumulate misfolded proteins, and proteotoxicity is involved in cadmium toxicity. To clear the roles of Akt in cadmium exposure-induced RTCs death, we investigated the possibility that Akt could regulate proteotoxicity through autophagy in cadmium chloride (CdCl2)-exposed HK-2 human renal proximal tubular cells. CdCl2 exposure promoted the accumulation of misfolded or damaged proteins, the formation of aggresomes (pericentriolar cytoplasmic inclusions), and aggrephagy (selective autophagy to degrade aggresome). Pharmacological inhibition of Akt using MK2206 or Akti-1/2 enhanced aggrephagy by promoting dephosphorylation and nuclear translocation of transcription factor EB (TFEB)/transcription factor E3 (TFE3), lysosomal transcription factors. TFEB or TFE3 knockdown by siRNAs attenuated the protective effects of MK2206 against cadmium toxicity. These results suggested that aberrant activation of Akt attenuates aggrephagy via TFEB or TFE3 to facilitate CdCl2-induced cell death. Furthermore, these roles of Akt/TFEB/TFE3 were conserved in CdCl2-exposed primary human RTCs. The present study shows the molecular mechanisms underlying Akt activation that promotes cadmium-induced RTCs death.


Sujet(s)
Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Cadmium , Protéines proto-oncogènes c-akt , Humains , Protéines proto-oncogènes c-akt/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Lignée cellulaire , Cadmium/toxicité , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Phosphorylation/effets des médicaments et des substances chimiques , Chlorure de cadmium/toxicité , Composés hétérocycliques 3 noyaux/pharmacologie , Tubules rénaux/métabolisme , Tubules rénaux/effets des médicaments et des substances chimiques , Tubules rénaux/cytologie , Tubules rénaux/anatomopathologie
5.
Exp Cell Res ; 440(1): 114116, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38830568

RÉSUMÉ

During the progression of diabetic kidney disease, proximal tubular epithelial cells respond to high glucose to induce hypertrophy and matrix expansion leading to renal fibrosis. Recently, a non-canonical PTEN has been shown to be translated from an upstream initiation codon CUG (leucine) to produce a longer protein called PTEN-Long (PTEN-L). Interestingly, the extended sequence present in PTEN-L contains cell secretion/penetration signal. Role of this non-canonical PTEN-L in diabetic renal tubular injury is not known. We show that high glucose decreases expression of PTEN-L. As a mechanism of its function, we find that reduced PTEN-L activates Akt-2, which phosphorylates and inactivate tuberin and PRAS40, resulting in activation of mTORC1 in tubular cells. Antibacterial agent acriflavine and antiviral agent ATA regulate translation from CUG codon. Acriflavine and ATA, respectively, decreased and increased expression of PTEN-L to altering Akt-2 and mTORC1 activation in the absence of change in expression of canonical PTEN. Consequently, acriflavine and ATA modulated high glucose-induced tubular cell hypertrophy and lamininγ1 expression. Importantly, expression of PTEN-L inhibited high glucose-stimulated Akt/mTORC1 activity to abrogate these processes. Since PTEN-L contains secretion/penetration signals, addition of conditioned medium containing PTEN-L blocked Akt-2/mTORC1 activity. Notably, in renal cortex of diabetic mice, we found reduced PTEN-L concomitant with Akt-2/mTORC1 activation, leading to renal hypertrophy and lamininγ1 expression. These results present first evidence for involvement of PTEN-L in diabetic kidney disease.


Sujet(s)
Néphropathies diabétiques , Glucose , Tubules contournés proximaux , Complexe-1 cible mécanistique de la rapamycine , Phosphohydrolase PTEN , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/génétique , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Glucose/métabolisme , Glucose/pharmacologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Régulation négative/effets des médicaments et des substances chimiques , Souris , Humains , Mâle , Souris de lignée C57BL , Transduction du signal
6.
Ren Fail ; 46(2): 2359638, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38832484

RÉSUMÉ

Emerging data have revealed that damage to tubular epithelial cell is a driving force in the progression of diabetic kidney disease (DKD). However, the specific mechanisms by which lipotoxicity contributes to the injury of these cells, thereby influencing the development of DKD, are yet to be fully understood. Here, we analyzed the GSE 30529 microarray datasets of human tubulointerstitial tissue samples from the Gene Expression Omnibus database (GEO). Concurrently, we conducted RNA-sequencing on palmitic acid (PA)-treated human renal proximal tubule epithelial cells (HK2 cells). After normalization, the differentially expressed genes (DEGs) were screened by R software and gene ontology (GO) enrichment analysis was conducted, and lysosomal-associated protein transmembrane 5 (LAPTM5) was finally selected. Our findings indicate that the expression of LAPTM5 was obviously increased in DKD patients, and the correlation between LAPTM5, and other clinical parameters of DKD was analyzed using the Spearman correlation analysis. The potential of LAPTM5 as a prognostic biomarker for DKD was further consolidated through receiver operating characteristic (ROC) analysis. To further verify the function of LAPTM5, we established mouse or in vitro systems mimicking DKD. The results showed that a consistent upregulation of LAPTM5, which was also found to be linked with inflammatory mediators within the context of DKD. Additionally, LAPTM5 silencing significantly downregulated mRNA expression of inflammatory factors in PA-treated HK2 cells. These results indicate that LAPTM5 is a potential biomarker and therapeutic treatment target for DKD. This discovery paves the way for future research and development of targeted interventions aimed at mitigating the progression of this prevalent condition.


Sujet(s)
Biologie informatique , Néphropathies diabétiques , Protéines membranaires , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/génétique , Néphropathies diabétiques/étiologie , Néphropathies diabétiques/anatomopathologie , Humains , Animaux , Souris , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Lignée cellulaire , Acide palmitique/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Régulation positive , Marqueurs biologiques/métabolisme
7.
Ren Fail ; 46(1): 2347462, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38832497

RÉSUMÉ

Diabetic nephropathy (DN) is one of the most serious and frequent complications among diabetes patients and presently constitutes vast the cases of end-stage renal disease worldwide. Tubulointerstitial fibrosis is a crucial factor related to the occurrence and progression of DN. Oridonin (Ori) is a diterpenoid derived from rubescens that has diverse pharmacological properties. Our previous study showed that Ori can protect against DN by decreasing the inflammatory response. However, whether Ori can alleviate renal fibrosis in DN remains unknown. Here, we investigated the mechanism through which Ori affects the Wnt/ß-catenin signaling pathway in diabetic rats and human proximal tubular epithelial cells (HK-2) exposed to high glucose (HG) levels. Our results revealed that Ori treatment markedly decreased urinary protein excretion levels, improved renal function and alleviated renal fibrosis in diabetic rats. In vitro, HG treatment increased the migration of HK-2 cells while reducing their viability and proliferation rate, and treatment with Ori reversed these changes. Additionally, the knockdown of ß-catenin arrested cell migration and reduced the expression levels of Wnt/ß-catenin signaling-related molecules (Wnt4, p-GSK3ß and ß-catenin) and fibrosis-related molecules (α-smooth muscle actin, collagen I and fibronectin), and Ori treatment exerted an effect similar to that observed after the knockdown of ß-catenin. Furthermore, the combination of Ori treatment and ß-catenin downregulation exerted more pronounced biological effects than treatment alone. These findings may provide the first line of evidence showing that Ori alleviates fibrosis in DN by inhibiting the Wnt/ß-catenin signaling pathway and thereby reveal a novel therapeutic avenue for treating tubulointerstitial fibrosis.


Sujet(s)
Diabète expérimental , Néphropathies diabétiques , Diterpènes de type kaurane , Fibrose , Rat Sprague-Dawley , Voie de signalisation Wnt , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/étiologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Animaux , Diterpènes de type kaurane/pharmacologie , Diterpènes de type kaurane/usage thérapeutique , Rats , Fibrose/traitement médicamenteux , Humains , Mâle , Diabète expérimental/complications , Diabète expérimental/traitement médicamenteux , Lignée cellulaire , bêta-Caténine/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme
8.
Sci Rep ; 14(1): 14004, 2024 06 18.
Article de Anglais | MEDLINE | ID: mdl-38890434

RÉSUMÉ

Cisplatin is an effective chemotherapeutic agent widely used for the treatment of various solid tumors. However, cisplatin has an important limitation in its use; currently, there is no method to ameliorate cisplatin-induced acute kidney injury (AKI). Thrombomodulin (TM) is well known not only for its role as a cofactor in the clinically important natural anticoagulation pathway but also for its anti-inflammatory properties. Here, we investigated the effects of TM in cisplatin-induced AKI. In mice intraperitoneally injected with 15 mg/kg cisplatin, TM (10 mg/kg) or PBS was administered intravenously at 24 h after cisplatin injection. TM significantly attenuated cisplatin-induced nephrotoxicity with the suppressed elevation of blood urea nitrogen and serum creatinine, and reduced histological damages. Actually, TM treatment significantly alleviated oxidative stress-induced apoptosis by reducing reactive oxygen species (ROS) levels in cisplatin-treated renal proximal tubular epithelial cells (RPTECs) in vitro. Furthermore, TM clarified cisplatin-induced apoptosis by reducing caspase-3 levels. In addition, TM attenuated the endoplasmic reticulum (ER) stress signaling pathway in both renal tissues and RPTECs to protect the kidneys from cisplatin-induced AKI. These findings suggest that TM is a potential protectant against cisplatin-induced nephrotoxicity through suppressing ROS generation and ER stress in response to cisplatin.


Sujet(s)
Atteinte rénale aigüe , Apoptose , Cisplatine , Stress du réticulum endoplasmique , Stress oxydatif , Espèces réactives de l'oxygène , Thrombomoduline , Cisplatine/effets indésirables , Animaux , Thrombomoduline/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/anatomopathologie , Souris , Espèces réactives de l'oxygène/métabolisme , Mâle , Apoptose/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Antinéoplasiques/effets indésirables , Antinéoplasiques/toxicité , Souris de lignée C57BL , Azote uréique sanguin , Transduction du signal/effets des médicaments et des substances chimiques , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie
9.
Cell Death Dis ; 15(6): 397, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844455

RÉSUMÉ

Integrin αvß6 holds promise as a therapeutic target for organ fibrosis, yet targeted therapies are hampered by concerns over inflammatory-related side effects. The role of αvß6 in renal inflammation remains unknown, and clarifying this issue is crucial for αvß6-targeted treatment of chronic kidney disease (CKD). Here, we revealed a remarkable positive correlation between overexpressed αvß6 in proximal tubule cells (PTCs) and renal inflammation in CKD patients and mouse models. Notably, knockout of αvß6 not only significantly alleviated renal fibrosis but also reduced inflammatory responses in mice, especially the infiltration of pro-inflammatory macrophages. Furthermore, conditional knockout of αvß6 in PTCs in vivo and co-culture of PTCs with macrophages in vitro showed that depleting αvß6 in PTCs suppressed the migration and pro-inflammatory differentiation of macrophages. Screening of macrophage activators showed that αvß6 in PTCs activates macrophages via secreting IL-34. IL-34 produced by PTCs was significantly diminished by αvß6 silencing, and reintroduction of IL-34 restored macrophage activities, while anti-IL-34 antibody restrained macrophage activities enhanced by αvß6 overexpression. Moreover, RNA-sequencing of PTCs and verification experiments demonstrated that silencing αvß6 in PTCs blocked hypoxia-stimulated IL-34 upregulation and secretion by inhibiting YAP expression, dephosphorylation, and nuclear translocation, which resulted in the activation of Hippo signaling. While application of a YAP agonist effectively recurred IL-34 production by PTCs, enhancing the subsequent macrophage migration and activation. Besides, reduced IL-34 expression and YAP activation were also observed in global or PTCs-specific αvß6-deficient injured kidneys. Collectively, our research elucidates the pro-inflammatory function and YAP/IL-34/macrophage axis-mediated mechanism of αvß6 in renal inflammation, providing a solid rationale for the use of αvß6 inhibition to treat kidney inflammation and fibrosis.


Sujet(s)
Intégrines , Macrophages , Souris knockout , Insuffisance rénale chronique , Animaux , Macrophages/métabolisme , Souris , Humains , Intégrines/métabolisme , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Inflammation/anatomopathologie , Inflammation/métabolisme , Mâle , Antigènes néoplasiques/métabolisme , Souris de lignée C57BL , Transduction du signal , Modèles animaux de maladie humaine , Protéines de signalisation YAP/métabolisme , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Fibrose
10.
Kidney Int ; 106(1): 16-18, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38906649

RÉSUMÉ

Glomerular issues and affected podocytes are at the origin of 80% of chronic kidney disease cases. Thus, acquiring a deeper understanding in this domain is necessary to halt progressive kidney damage. In this study, the authors investigated the harmful impact of podocyte-cleaved soluble retinoic acid receptor responder protein-1 on podocytes and proximal tubular cells and identified matrix metalloprotease 23 as the enzyme responsible for cleaving retinoic acid receptor responder protein-1. These findings provide new insights into chronic kidney disease progression, suggesting innovative treatment avenues.


Sujet(s)
Évolution de la maladie , Podocytes , Insuffisance rénale chronique , Podocytes/métabolisme , Podocytes/anatomopathologie , Podocytes/effets des médicaments et des substances chimiques , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Humains , Animaux , Souris , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Lignée cellulaire , Protéolyse
11.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892409

RÉSUMÉ

Renal ischemia/reperfusion is a serious condition that not only causes acute kidney injury, a severe clinical syndrome with high mortality, but is also an inevitable part of kidney transplantation or other kidney surgeries. Alterations of oxygen levels during ischemia/reperfusion, namely hypoxia/reoxygenation, disrupt mitochondrial metabolism and induce structural changes that lead to cell death. A signature mitochondrial phospholipid, cardiolipin, with many vital roles in mitochondrial homeostasis, is one of the key players in hypoxia/reoxygenation-induced mitochondrial damage. In this study, we analyze the effect of hypoxia/reoxygenation on human renal proximal tubule epithelial cell (RPTEC) cardiolipins, as well as their metabolism and mitochondrial functions. RPTEC cells were placed in a hypoxic chamber with a 2% oxygen atmosphere for 24 h to induce hypoxia; then, they were replaced back into regular growth conditions for 24 h of reoxygenation. Surprisingly, after 24 h, hypoxia cardiolipin levels substantially increased and remained higher than control levels after 24 h of reoxygenation. This was explained by significantly elevated levels of cardiolipin synthase and lysocardiolipin acyltransferase 1 (LCLAT1) gene expression and protein levels. Meanwhile, hypoxia/reoxygenation decreased ADP-dependent mitochondrial respiration rates and oxidative phosphorylation capacity and increased reactive oxygen species generation. Our findings suggest that hypoxia/reoxygenation induces cardiolipin remodeling in response to reduced mitochondrial oxidative phosphorylation in a way that protects mitochondrial function.


Sujet(s)
Cardiolipides , Hypoxie cellulaire , Mitochondries , Oxygène , Espèces réactives de l'oxygène , Humains , Cardiolipides/métabolisme , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Oxygène/métabolisme , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/cytologie , Phosphorylation oxydative , Rein/métabolisme , Rein/anatomopathologie , Lignée cellulaire , Transferases (other substituted phosphate groups)/métabolisme , Transferases (other substituted phosphate groups)/génétique , Protéines membranaires
12.
Kidney Int ; 106(1): 50-66, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38697478

RÉSUMÉ

Retinoic acid receptor responder protein-1 (RARRES1) is a podocyte-enriched transmembrane protein whose increased expression correlates with human glomerular disease progression. RARRES1 promotes podocytopenia and glomerulosclerosis via p53-mediated podocyte apoptosis. Importantly, the cytopathic actions of RARRES1 are entirely dependent on its proteolytic cleavage into a soluble protein (sRARRES1) and subsequent podocyte uptake by endocytosis, as a cleavage mutant RARRES1 exerted no effects in vitro or in vivo. As RARRES1 expression is upregulated in human glomerular diseases, here we investigated the functional consequence of podocyte-specific overexpression of RARRES1 in mice in the experimental focal segmental glomerulosclerosis and diabetic kidney disease. We also examined the effects of long-term RARRES1 overexpression on slowly developing aging-induced kidney injury. As anticipated, the induction of podocyte overexpression of RARRES1 (Pod-RARRES1WT) significantly worsened glomerular injuries and worsened kidney function in all three models, while overexpression of RARRES1 cleavage mutant (Pod-RARRES1MT) did not. Remarkably, direct uptake of sRARRES1 was also seen in proximal tubules of injured Pod-RARRES1WT mice and associated with exacerbated tubular injuries, vacuolation, and lipid accumulation. Single-cell RNA sequence analysis of mouse kidneys demonstrated RARRES1 led to a marked deregulation of lipid metabolism in proximal tubule subsets. We further identified matrix metalloproteinase 23 (MMP23) as a highly podocyte-specific metalloproteinase and responsible for RARRES1 cleavage in disease settings, as adeno-associated virus 9-mediated knockdown of MMP23 abrogated sRARRES1 uptake in tubular cells in vivo. Thus, our study delineates a previously unrecognized mechanism by which a podocyte-derived protein directly facilitates podocyte and tubular injury in glomerular diseases and suggests that podocyte-specific functions of RARRES1 and MMP23 may be targeted to ameliorate glomerular disease progression in vivo.


Sujet(s)
Néphropathies diabétiques , Évolution de la maladie , Glomérulonéphrite segmentaire et focale , Tubules contournés proximaux , Podocytes , Podocytes/métabolisme , Podocytes/anatomopathologie , Animaux , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/génétique , Néphropathies diabétiques/étiologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Humains , Glomérulonéphrite segmentaire et focale/anatomopathologie , Glomérulonéphrite segmentaire et focale/métabolisme , Glomérulonéphrite segmentaire et focale/génétique , Souris , Modèles animaux de maladie humaine , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Mâle , Souris de lignée C57BL , Souris transgéniques , Apoptose , Endocytose
13.
Am J Physiol Renal Physiol ; 327(1): F137-F145, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38779756

RÉSUMÉ

Polymyxins are a last-resort treatment option for multidrug-resistant gram-negative bacterial infections, but they are associated with nephrotoxicity. Gelofusine was previously shown to reduce polymyxin-associated kidney injury in an animal model. However, the mechanism(s) of renal protection has not been fully elucidated. Here, we report the use of a cell culture model to provide insights into the mechanisms of renal protection. Murine epithelial proximal tubular cells were exposed to polymyxin B. Cell viability, lactate dehydrogenase (LDH) release, polymyxin B uptake, mitochondrial superoxide production, nuclear morphology, and apoptosis activation were evaluated with or without concomitant gelofusine. A megalin knockout cell line was used as an uptake inhibition control. Methionine was included in selected experiments as an antioxidant control. A polymyxin B concentration-dependent reduction in cell viability was observed. Increased viability was observed in megalin knockout cells following comparable polymyxin B exposures. Compared with polymyxin B exposure alone, concomitant gelofusine significantly increased cell viability as well as reduced LDH release, polymyxin B uptake, mitochondrial superoxide, and apoptosis. Gelofusine and methionine were more effective at reducing renal cell injury in combination than either agent alone. In conclusion, the mechanisms of renal protection by gelofusine involve decreasing cellular drug uptake, reducing subsequent oxidative stress and apoptosis activation. These findings would be valuable for translational research into clinical strategies to attenuate drug-associated acute kidney injury.NEW & NOTEWORTHY Gelofusine is a gelatinous saline solution with the potential to attenuate polymyxin-associated nephrotoxicity. We demonstrated that the mechanisms of gelofusine renal protection involve reducing polymyxin B uptake by proximal tubule cells, limiting subsequent oxidative stress and apoptosis activation. In addition, gelofusine was more effective at reducing cellular injury than a known antioxidant control, methionine, and a megalin knockout cell line, indicating that gelofusine likely has additional pharmacological properties besides only megalin inhibition.


Sujet(s)
Antibactériens , Apoptose , Polymyxine B , Animaux , Polymyxine B/pharmacologie , Souris , Apoptose/effets des médicaments et des substances chimiques , Antibactériens/pharmacologie , Antibactériens/toxicité , Survie cellulaire/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Lignée cellulaire , Protéine-2 apparentée au récepteur des LDL/métabolisme , Protéine-2 apparentée au récepteur des LDL/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/prévention et contrôle , Atteinte rénale aigüe/induit chimiquement , Stress oxydatif/effets des médicaments et des substances chimiques , L-Lactate dehydrogenase/métabolisme
14.
Am J Physiol Renal Physiol ; 327(1): F128-F136, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38695076

RÉSUMÉ

Acute kidney injury (AKI) is extremely prevalent among hospitalizations and presents a significant risk for the development of chronic kidney disease and increased mortality. Ischemia caused by shock, trauma, and transplant are common causes of AKI. To attenuate ischemic AKI therapeutically, we need a better understanding of the physiological and cellular mechanisms underlying damage. Instances of ischemia are most damaging in proximal tubule epithelial cells (PTECs) where hypoxic signaling cascades, and perhaps more rapidly, posttranslational modifications (PTMs), act in concert to change cellular metabolism. Here, we focus on the effects of the understudied PTM, lysine succinylation. We have previously shown a protective effect of protein hypersuccinylation on PTECs after depletion of the desuccinylase sirtuin5. General trends in the results suggested that hypersuccinylation led to upregulation of peroxisomal activity and was protective against kidney injury. Included in the list of changes was the Parkinson's-related deglycase Park7. There is little known about any links between peroxisome activity and Park7. In this study, we show in vitro and in vivo that Park7 has a crucial role in protection from AKI and upregulated peroxisome activity. These data in combination with published results of Park7's protective role in cardiovascular damage and chronic kidney disease lead us to hypothesize that succinylation of Park7 may ameliorate oxidative damage resulting from AKI and prevent disease progression. This novel mechanism provides a potential therapeutic mechanism that can be targeted.NEW & NOTEWORTHY Succinylation is an understudied posttranslational modification that has been shown to increase peroxisomal activity. Furthermore, increased peroxisomal activity has been shown to reduce oxidative stress and protect proximal tubules after acute kidney injury. Analysis of mass spectrometry succinylomic and proteomic data reveals a novel role for Parkinson's related Park7 in mediating Nrf2 antioxidant response after kidney injury. This novel protection pathway provides new insights for kidney injury prevention and development of novel therapeutics.


Sujet(s)
Atteinte rénale aigüe , Tubules contournés proximaux , Protein deglycase DJ-1 , Animaux , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/prévention et contrôle , Atteinte rénale aigüe/anatomopathologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Protein deglycase DJ-1/métabolisme , Protein deglycase DJ-1/génétique , Maturation post-traductionnelle des protéines , Souris de lignée C57BL , Modèles animaux de maladie humaine , Mâle , Sirtuines/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Transduction du signal , Souris , Stress oxydatif , Lysine/métabolisme
15.
J Pathol ; 263(3): 315-327, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38721910

RÉSUMÉ

Hemolysis-induced acute kidney injury (AKI) is attributed to heme-mediated proximal tubule epithelial cell (PTEC) injury and tubular cast formation due to intratubular protein condensation. Megalin is a multiligand endocytic receptor for proteins, peptides, and drugs in PTECs and mediates the uptake of free hemoglobin and the heme-scavenging protein α1-microglobulin. However, understanding of how megalin is involved in the development of hemolysis-induced AKI remains elusive. Here, we investigated the megalin-related pathogenesis of hemolysis-induced AKI and a therapeutic strategy using cilastatin, a megalin blocker. A phenylhydrazine-induced hemolysis model developed in kidney-specific mosaic megalin knockout (MegKO) mice confirmed megalin-dependent PTEC injury revealed by the co-expression of kidney injury molecule-1 (KIM-1). In the hemolysis model in kidney-specific conditional MegKO mice, the uptake of hemoglobin and α1-microglobulin as well as KIM-1 expression in PTECs was suppressed, but tubular cast formation was augmented, likely due to the nonselective inhibition of protein reabsorption in PTECs. Quartz crystal microbalance analysis revealed that cilastatin suppressed the binding of megalin with hemoglobin and α1-microglobulin. Cilastatin also inhibited the specific uptake of fluorescent hemoglobin by megalin-expressing rat yolk sac tumor-derived L2 cells. In a mouse model of hemolysis-induced AKI, repeated cilastatin administration suppressed PTEC injury by inhibiting the uptake of hemoglobin and α1-microglobulin and also prevented cast formation. Hemopexin, another heme-scavenging protein, was also found to be a novel ligand of megalin, and its binding to megalin and uptake by PTECs in the hemolysis model were suppressed by cilastatin. Mass spectrometry-based semiquantitative analysis of urinary proteins in cilastatin-treated C57BL/6J mice indicated that cilastatin suppressed the reabsorption of a limited number of megalin ligands in PTECs, including α1-microglobulin and hemopexin. Collectively, cilastatin-mediated selective megalin blockade is an effective therapeutic strategy to prevent both heme-mediated PTEC injury and cast formation in hemolysis-induced AKI. © 2024 The Pathological Society of Great Britain and Ireland.


Sujet(s)
Atteinte rénale aigüe , Hémolyse , Tubules contournés proximaux , Protéine-2 apparentée au récepteur des LDL , Souris knockout , Animaux , Protéine-2 apparentée au récepteur des LDL/métabolisme , Protéine-2 apparentée au récepteur des LDL/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Hémoglobines/métabolisme , Souris , Cilastatine/pharmacologie , Modèles animaux de maladie humaine , Phénylhydrazines , Souris de lignée C57BL , Mâle , Récepteur cellulaire-1 du virus de l'hépatite A/métabolisme , alpha-Globulines/métabolisme , Humains
16.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731829

RÉSUMÉ

Kidney ischemia and reperfusion injury (IRI) is a significant contributor to acute kidney injury (AKI), characterized by tubular injury and kidney dysfunction. Salvador family WW domain containing protein 1 (SAV1) is a key component of the Hippo pathway and plays a crucial role in the regulation of organ size and tissue regeneration. However, whether SAV1 plays a role in kidney IRI is not investigated. In this study, we investigated the role of SAV1 in kidney injury and regeneration following IRI. A proximal tubule-specific knockout of SAV1 in kidneys (SAV1ptKO) was generated, and wild-type and SAV1ptKO mice underwent kidney IRI or sham operation. Plasma creatinine and blood urea nitrogen were measured to assess kidney function. Histological studies, including periodic acid-Schiff staining and immunohistochemistry, were conducted to assess tubular injury, SAV1 expression, and cell proliferation. Western blot analysis was employed to assess the Hippo pathway-related and proliferation-related proteins. SAV1 exhibited faint expression in the proximal tubules and was predominantly expressed in the connecting tubule to the collecting duct. At 48 h after IRI, SAV1ptKO mice continued to exhibit severe kidney dysfunction, compared to attenuated kidney dysfunction in wild-type mice. Consistent with the functional data, severe tubular damage induced by kidney IRI in the cortex was significantly decreased in wild-type mice at 48 h after IRI but not in SAV1ptKO mice. Furthermore, 48 h after IRI, the number of Ki67-positive cells in the cortex was significantly higher in wild-type mice than SAV1ptKO mice. After IRI, activation and expression of Hippo pathway-related proteins were enhanced, with no significant differences observed between wild-type and SAV1ptKO mice. Notably, at 48 h after IRI, protein kinase B activation (AKT) was significantly enhanced in SAV1ptKO mice compared to wild-type mice. This study demonstrates that SAV1 deficiency in the kidney proximal tubule worsens the injury and delays kidney regeneration after IRI, potentially through the overactivation of AKT.


Sujet(s)
Atteinte rénale aigüe , Protéines du cycle cellulaire , Tubules contournés proximaux , Lésion d'ischémie-reperfusion , Animaux , Mâle , Souris , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/étiologie , Atteinte rénale aigüe/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Prolifération cellulaire , Modèles animaux de maladie humaine , Voie de signalisation Hippo , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Souris de lignée C57BL , Souris knockout , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/génétique , Transduction du signal
17.
Cell Mol Life Sci ; 81(1): 244, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38814462

RÉSUMÉ

Four-and-a-half LIM domains protein 2 (FHL2) is an adaptor protein that may interact with hypoxia inducible factor 1α (HIF-1α) or ß-catenin, two pivotal protective signaling in acute kidney injury (AKI). However, little is known about the regulation and function of FHL2 during AKI. We found that FHL2 was induced in renal tubular cells in patients with acute tubular necrosis and mice model of ischemia-reperfusion injury (IRI). In cultured renal proximal tubular cells (PTCs), hypoxia induced FHL2 expression and promoted the binding of HIF-1 to FHL2 promoter. Compared with control littermates, mice with PTC-specific deletion of FHL2 gene displayed worse renal function, more severe morphologic lesion, more tubular cell death and less cell proliferation, accompanying by downregulation of AQP1 and Na, K-ATPase after IRI. Consistently, loss of FHL2 in PTCs restricted activation of HIF-1 and ß-catenin signaling simultaneously, leading to attenuation of glycolysis, upregulation of apoptosis-related proteins and downregulation of proliferation-related proteins during IRI. In vitro, knockdown of FHL2 suppressed hypoxia-induced activation of HIF-1α and ß-catenin signaling pathways. Overexpression of FHL2 induced physical interactions between FHL2 and HIF-1α, ß-catenin, GSK-3ß or p300, and the combination of these interactions favored the stabilization and nuclear translocation of HIF-1α and ß-catenin, enhancing their mediated gene transcription. Collectively, these findings identify FHL2 as a direct downstream target gene of HIF-1 signaling and demonstrate that FHL2 could play a critical role in protecting against ischemic AKI by promoting the activation of HIF-1 and ß-catenin signaling through the interactions with its multiple protein partners.


Sujet(s)
Atteinte rénale aigüe , Tubules contournés proximaux , Protéines à homéodomaine LIM , Protéines du muscle , Lésion d'ischémie-reperfusion , Facteurs de transcription , bêta-Caténine , Animaux , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/génétique , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/génétique , Humains , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/génétique , Souris , bêta-Caténine/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Mâle , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Transduction du signal , Souris de lignée C57BL , Souris knockout , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , Prolifération cellulaire , Apoptose
18.
Mol Biol Rep ; 51(1): 620, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38709349

RÉSUMÉ

BACKGROUND: Recent years of evidence suggest the crucial role of renal tubular cells in developing diabetic kidney disease. Scopoletin (SCOP) is a plant-based coumarin with numerous biological activities. This study aimed to determine the effect of SCOP on renal tubular cells in developing diabetic kidney disease and to elucidate mechanisms. METHODS AND RESULTS: In this study, SCOP was evaluated in vitro using renal proximal tubular (HK-2) cells under hyperglycemic conditions to understand its mechanism of action. In HK-2 cells, SCOP alleviated the high glucose-generated reactive oxygen species (ROS), restored the levels of reduced glutathione, and decreased lipid peroxidation. High glucose-induced alteration in the mitochondrial membrane potential was markedly restored in the SCOP-treated cells. Moreover, SCOP significantly reduced the high glucose-induced apoptotic cell population in the Annexin V-FITC flow cytometry study. Furthermore, high glucose markedly elevated the mRNA expression of fibrotic and extracellular matrix (ECM) components, namely, transforming growth factor (TGF)-ß, alfa-smooth muscle actin (α-SMA), collagen I, and collagen III, in HK-2 cells compared to the untreated cells. SCOP treatment reduced these mRNA expressions compared to the high glucose-treated cells. Collagen I and TGF-ß protein levels were also significantly reduced in the SCOP-treated cells. Further findings in HK-2 cells revealed that SCOP interfered with the epithelial-mesenchymal transition (EMT) in the high glucose-treated HK-2 cells by normalizing E-cadherin and downregulating the vimentin and α-SMA proteins. CONCLUSIONS: In conclusion, SCOP modulates the high glucose-generated renal tubular cell oxidative damage and accumulation of ECM components and may be a promising molecule against diabetic nephropathy.


Sujet(s)
Néphropathies diabétiques , Transition épithélio-mésenchymateuse , Glucose , Tubules contournés proximaux , Stress oxydatif , Espèces réactives de l'oxygène , Scopolétine , Humains , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Glucose/métabolisme , Glucose/pharmacologie , Glucose/toxicité , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Scopolétine/pharmacologie , Lignée cellulaire , Espèces réactives de l'oxygène/métabolisme , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Fibrose , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques
19.
FASEB J ; 38(10): e23688, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38780519

RÉSUMÉ

Diabetic nephropathy (DN) is a major cause of chronic kidney disease. Microalbuminuria is currently the most common non-invasive biomarker for the early diagnosis of DN. However, renal structural damage may have advanced when albuminuria is detected. In this study, we sought biomarkers for early DN diagnosis through proteomic analysis of urinary extracellular vesicles (uEVs) from type 2 diabetic model rats and normal controls. Isocitrate dehydrogenase 1 (IDH1) was significantly increased in uEVs from diabetic model rats at the early stage despite minimal differences in albuminuria between the groups. Calorie restriction significantly suppressed the increase in IDH1 in uEVs and 24-hour urinary albumin excretion, suggesting that the increase in IDH1 in uEVs was associated with the progression of DN. Additionally, we investigated the origin of IDH1-containing uEVs based on their surface sugar chains. Lectin affinity enrichment and immunohistochemical staining showed that IDH1-containing uEVs were derived from proximal tubules. These findings suggest that the increase in IDH1 in uEVs reflects pathophysiological alterations in the proximal tubules and that IDH1 in uEVs may serve as a potential biomarker of DN in the proximal tubules.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Néphropathies diabétiques , Vésicules extracellulaires , Isocitrate dehydrogenases , Tubules contournés proximaux , Régulation positive , Animaux , Isocitrate dehydrogenases/métabolisme , Isocitrate dehydrogenases/génétique , Vésicules extracellulaires/métabolisme , Rats , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Diabète de type 2/urine , Diabète de type 2/métabolisme , Mâle , Néphropathies diabétiques/urine , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Diabète expérimental/métabolisme , Diabète expérimental/urine , Rat Sprague-Dawley , Marqueurs biologiques/urine , Marqueurs biologiques/métabolisme
20.
Hum Cell ; 37(4): 1039-1055, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38753279

RÉSUMÉ

The link between ferroptosis, a form of cell death mediated by iron and acute kidney injury (AKI) is recently gaining widespread attention. However, the mechanism of the crosstalk between cells in the pathogenesis and progression of acute kidney injury remains unexplored. In our research, we performed a non-negative matrix decomposition (NMF) algorithm on acute kidney injury single-cell RNA sequencing data based specifically focusing in ferroptosis-associated genes. Through a combination with pseudo-time analysis, cell-cell interaction analysis and SCENIC analysis, we discovered that proximal tubular cells, macrophages, and fibroblasts all showed associations with ferroptosis in different pathways and at various time. This involvement influenced cellular functions, enhancing cellular communication and activating multiple transcription factors. In addition, analyzing bulk expression profiles and marker genes of newly defined ferroptosis subtypes of cells, we have identified crucial cell subtypes, including Egr1 + PTC-C1, Jun + PTC-C3, Cxcl2 + Mac-C1 and Egr1 + Fib-C1. All these subtypes which were found in AKI mice kidneys and played significantly distinct roles from those of normal mice. Moreover, we verified the differential expression of Egr1, Jun, and Cxcl2 in the IRI mouse model and acute kidney injury human samples. Finally, our research presented a novel analysis of the crosstalk of proximal tubular cells, macrophages and fibroblasts in acute kidney injury targeting ferroptosis, therefore, contributing to better understanding the acute kidney injury pathogenesis, self-repairment and acute kidney injury-chronic kidney disease (AKI-CKD) progression.


Sujet(s)
Atteinte rénale aigüe , Ferroptose , Fibroblastes , Tubules contournés proximaux , Macrophages , Analyse sur cellule unique , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/métabolisme , Ferroptose/génétique , Ferroptose/physiologie , Macrophages/métabolisme , Macrophages/physiologie , Humains , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Analyse sur cellule unique/méthodes , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/cytologie , Souris , Communication cellulaire , Modèles animaux de maladie humaine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...