Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.505
Filtrer
1.
Sci Rep ; 14(1): 22927, 2024 10 02.
Article de Anglais | MEDLINE | ID: mdl-39358473

RÉSUMÉ

Primary cultures of proximal tubule cells are widely used to model the behavior of kidney epithelial cells in vitro. However, de-differentiation of primary cells upon culture has been observed and appreciated for decades, yet the mechanisms driving this phenomenon remain poorly understood. This confounds the interpretation of experiments using primary kidney epithelial cells and prevents their use to engineer functional kidney tissue ex vivo. In this report, we measure the dynamics of cell-state transformations in early primary culture of mouse proximal tubules to identify key pathways and processes that correlate with and may drive de-differentiation. Our data show that the loss of proximal-tubule-specific genes is rapid, uniform, and sustained even after confluent, polarized epithelial monolayers develop. This de-differentiation occurs uniformly across many common culture condition variations. Changes in early culture were strongly associated with the loss of HNF4A. Exogenous re-expression of HNF4A can promote expression of a subset of proximal tubule genes in a de-differentiated proximal tubule cell line. Using genetically labeled proximal tubule cells, we show that selective pressures very early in culture influence which cells grow to confluence. Together, these data indicate that the loss of in vivo function in proximal tubule cultures occurs very early and suggest that the sustained loss of HNF4A is a key regulatory event mediating this change.


Sujet(s)
Facteur nucléaire hépatocytaire HNF-4 , Tubules contournés proximaux , Animaux , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/cytologie , Facteur nucléaire hépatocytaire HNF-4/génétique , Facteur nucléaire hépatocytaire HNF-4/métabolisme , Souris , Cellules épithéliales/métabolisme , Cellules cultivées , Différenciation cellulaire/génétique , Culture de cellules primaires , Régulation de l'expression des gènes , Lignée cellulaire
2.
Genes (Basel) ; 15(9)2024 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-39336766

RÉSUMÉ

Dent disease type 1 is a rare X-linked recessive inherited renal disorder affecting mainly young males, generally leading to end-stage renal failure and for which there is no cure. It is caused by inactivating mutations in the gene encoding ClC-5, a 2Cl-/H+ exchanger found on endosomes in the renal proximal tubule. This transporter participates in reabsorbing all filtered plasma proteins, which justifies why proteinuria is commonly observed when ClC-5 is defective. In the context of Dent disease type 1, a proximal tubule dedifferentiation was shown to be accompanied by a dysfunctional cell metabolism. However, the exact mechanisms linking such alterations to chronic kidney disease are still unclear. In this review, we gather knowledge from several Dent disease type 1 models to summarize the current hypotheses generated to understand the progression of this disorder. We also highlight some urinary biomarkers for Dent disease type 1 suggested in different studies.


Sujet(s)
Canaux chlorure , Tubules contournés proximaux , Humains , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/physiopathologie , Canaux chlorure/génétique , Canaux chlorure/métabolisme , Animaux , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/anatomopathologie , Maladies génétiques liées au chromosome X/physiopathologie , Mutation , Marqueurs biologiques/urine , Néphrolithiase
3.
Mol Biol Rep ; 51(1): 974, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39259342

RÉSUMÉ

BACKGROUND: One of the causes of tubulointerstitial nephritis is viral infection, with innate immune responses affecting its pathogenesis. Toll-like receptor 3 (TLR3) recognizes viral infections and acts antivirally by activating signaling to produce inflammatory cytokines/chemokines, including C-C motif chemokine ligand 5 (CCL5) and interferon-ß (IFN-ß). Although cylindromatosis lysine 63 deubiquitinase (CYLD) is known to be associated with tubulointerstitial nephritis and renal function, its role in the antiviral innate immune response in tubular epithelial cells remains unknown. In this study, we investigated the association between CYLD and TLR3-mediated CCL5 production in cultured human renal proximal tubular epithelial cells (hRPTECs). METHODS AND RESULTS: Polyinosinic-polycytidylic acid (poly IC), a synthetic TLR3 ligand, was used to stimulate hRPTECs. mRNA expression was measured using reverse transcription-quantitative polymerase chain reaction. Protein expression was assayed using western blotting or an enzyme-linked immunosorbent assay. Knockdown of IFN-ß, nuclear factor-kappa B (NF-κB) p65, and CYLD was performed by transfecting cells with specific small interfering RNAs. The intracellular localization of CYLD in hRPTECs was analyzed using immunofluorescence. Poly IC induced CCL5 expression in a time- and concentration-dependent manner, and knockdown of either IFN-ß or p65 reduced poly IC-induced CCL5 expression. CYLD knockdown increased the poly IC-induced CCL5, phosphorylated IκB kinase α/ß (IKK complex), and phosphorylated p65 expression. The CYLD protein was localized in the cytoplasm, and poly IC did not alter its expression. CONCLUSION: CYLD may prevent excessive inflammation due to an antiviral innate immune response by suppressing IKK complex and NF-κB activation downstream of TLR3 in hRPTECs.


Sujet(s)
Chimiokine CCL5 , Deubiquitinating enzyme CYLD , Cellules épithéliales , Tubules contournés proximaux , Poly I-C , Récepteur de type Toll-3 , Humains , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Deubiquitinating enzyme CYLD/métabolisme , Deubiquitinating enzyme CYLD/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Tubules contournés proximaux/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Poly I-C/pharmacologie , Interféron bêta/métabolisme , Interféron bêta/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription RelA/métabolisme , Immunité innée , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire
4.
J Biochem Mol Toxicol ; 38(10): e23854, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39287333

RÉSUMÉ

Ischemia-reperfusion (I-R) injury is the most common cause of acute kidney injury. In experiments involving primary human renal proximal tubular epithelial cells (RPTECs) exposed to anoxia-reoxygenation, we explored the hypothesis that mitochondrial malate dehydrogenase-2 (MDH-2) inhibition redirects malate metabolism from the mitochondria to the cytoplasm, towards the malate-pyruvate cycle and reversed malate-aspartate shuttle. Colorimetry, fluorometry, and western blotting showed that MDH2 inhibition accelerates the malate-pyruvate cycle enhancing cytoplasmic NADPH, thereby regenerating the potent antioxidant reduced glutathione. It also reversed the malate-aspartate shuttle and potentially diminished mitochondrial reactive oxygen species (ROS) production by transferring electrons, in the form of NADH, from the mitochondria to the cytoplasm. The excessive ROS production induced by anoxia-reoxygenation led to DNA damage and protein modification, triggering DNA damage and unfolded protein response, ultimately resulting in apoptosis and senescence. Additionally, ROS induced lipid peroxidation, which may contribute to the process of ferroptosis. Inhibiting MDH-2 proved effective in mitigating ROS overproduction during anoxia-reoxygenation, thereby rescuing RPTECs from death or senescence. Thus, targeting MDH-2 holds promise as a pharmaceutical strategy against I-R injury.


Sujet(s)
Cellules épithéliales , Malate dehydrogenase , Espèces réactives de l'oxygène , Espèces réactives de l'oxygène/métabolisme , Humains , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Malate dehydrogenase/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/cytologie , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Altération de l'ADN , Apoptose/effets des médicaments et des substances chimiques
5.
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
6.
Cell Mol Life Sci ; 81(1): 385, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39235496

RÉSUMÉ

Cisplatin-induced renal tubular injury largely restricts the wide-spread usage of cisplatin in the treatment of malignancies. Identifying the key signaling pathways that regulate cisplatin-induced renal tubular injury is thus clinically important. PARVB, a focal adhesion protein, plays a crucial role in tumorigenesis. However, the function of PARVB in kidney disease is largely unknown. To investigate whether and how PARVB contributes to cisplatin-induced renal tubular injury, a mouse model (PARVB cKO) was generated in which PARVB gene was specifically deleted from proximal tubular epithelial cells using the Cre-LoxP system. In this study, we found depletion of PARVB in proximal tubular epithelial cells significantly attenuates cisplatin-induced renal tubular injury, including tubular cell death and inflammation. Mechanistically, PARVB associates with transforming growth factor-ß-activated kinase 1 (TAK1), a central regulator of cell survival and inflammation that is critically involved in mediating cisplatin-induced renal tubular injury. Depletion of PARVB promotes cisplatin-induced TAK1 degradation, inhibits TAK1 downstream signaling, and ultimately alleviates cisplatin-induced tubular cell damage. Restoration of PARVB or TAK1 in PARVB-deficient cells aggravates cisplatin-induced tubular cell injury. Finally, we demonstrated that PARVB regulates TAK1 protein expression through an E3 ligase ITCH-dependent pathway. PARVB prevents ITCH association with TAK1 to block its ubiquitination. Our study reveals that PARVB deficiency protects against cisplatin-induced tubular injury through regulation of TAK1 signaling and indicates targeting this pathway may provide a novel therapeutic strategy to alleviate cisplatin-induced kidney damage.


Sujet(s)
Cisplatine , MAP Kinase Kinase Kinases , Souris knockout , Transduction du signal , Cisplatine/effets indésirables , Cisplatine/toxicité , Animaux , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Humains , Souris de lignée C57BL , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/effets indésirables , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Tubules rénaux/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal
7.
Nat Commun ; 15(1): 7010, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39237549

RÉSUMÉ

Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, together with injury-invoked inflammation and fibrosis. Deciphering the molecular pathways and cellular interactions driving these processes is challenging due to the complex tissue structure. Here, we apply single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics reveals injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicts Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and their neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis identifies cellular microenvironments resembling early tertiary lymphoid structures and associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.


Sujet(s)
Communication cellulaire , Microenvironnement cellulaire , Rein , Régénération , Lésion d'ischémie-reperfusion , Transcriptome , Animaux , Souris , Régénération/génétique , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/génétique , Lésion d'ischémie-reperfusion/anatomopathologie , Rein/métabolisme , Rein/anatomopathologie , Souris de lignée C57BL , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Mâle , Fibroblastes/métabolisme , Analyse de profil d'expression de gènes , Analyse sur cellule unique , Fibrose
8.
Biol Pharm Bull ; 47(9): 1557-1564, 2024.
Article de Anglais | MEDLINE | ID: mdl-39313392

RÉSUMÉ

Oxidative stress plays a crucial role in the development and progression of various kidney diseases. Nuclear factor erythroid 2-related factor 2 (NRF2) is the primary transcription factor that protects cells from oxidative stress by regulating cytoprotective genes including those involved in the antioxidant glutathione (GSH) pathway. GSH maintains cellular redox status and affects redox signaling, cell proliferation, and cell death. Antimycin A, an inhibitor of complex III of the electron transport chain, causes oxidative stress and reduces GSH levels. In this study, we induced mitochondrial damage in rat renal proximal tubular cells using antimycin A and investigated cellular viability and levels of NRF2 and GSH. Treatment with antimycin A altered the expression of antioxidant genes, including reduction in the transcription of glutathione-cysteine ligase subunits (Gclc and Gclm) and glutathione reductase (Gsr1), followed by a reduction in total GSH content with a concomitant decrease in NRF2 protein expression. AR-20007, previously described as an NRF2 activator, stabilizes and increases NRF2 protein expression in cells. By stimulating NRF2, AR-20007 increased the expression of antioxidant and detoxifying enzymes, thereby enhancing protection against oxidative stress induced by antimycin A. These data suggest that NRF2 activation effectively inhibits antimycin A-induced oxidative stress and that NRF2 may be a promising therapeutic target for preventing cell death during acute kidney injury.


Sujet(s)
Antimycine A , Cellules épithéliales , Glutathion , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Antimycine A/pharmacologie , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Glutathion/métabolisme , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Mort cellulaire/effets des médicaments et des substances chimiques , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Tubules rénaux/cytologie , Tubules rénaux/effets des médicaments et des substances chimiques , Tubules rénaux/métabolisme
9.
Antimicrob Agents Chemother ; 68(10): e0021924, 2024 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-39225483

RÉSUMÉ

This study aimed to assess the nephrotoxicity associated with VRP-034 (novel formulation of polymyxin B [PMB]) compared to marketed PMB in a three-dimensional (3D) kidney-on-a-chip model. To model the human kidney proximal tubule for analysis, tubular structures were established using 23 triple-channel chips seeded with RPTEC/hTERT1 cells. These cells were exposed to VRP-034 or PMB at seven concentrations (1-200 µM) over 12, 24, and 48 h. A suite of novel kidney injury biomarkers, cell health, and inflammatory markers were quantitatively assessed in the effluent. Additionally, caspase and cytochrome C levels were measured, and cell viability was evaluated using calcein AM and ethidium homodimer-1 (EthD-1). Exposure to marketed PMB resulted in significantly elevated levels (P < 0.05) of four key biomarkers (KIM-1, cystatin C, clusterin, and OPN) compared to VRP-034, particularly at clinically relevant concentrations of ≥10 µM. At 25 µM, all biomarkers demonstrated a significant increase (P < 0.05) with marketed PMB exposure compared to VRP-034. Inflammatory markers (interleukin-6 and interleukin-8) increased significantly (P < 0.05) with marketed PMB at concentrations of ≥5 µM, relative to VRP-034. VRP-034 displayed superior cell health outcomes, exhibiting lower lactate dehydrogenase release, while ATP levels remained comparable. Morphological analysis revealed that marketed PMB induced more severe damage, disrupting tubular integrity. Both treatments activated cytochrome C, caspase-3, caspase-8, caspase-9, and caspase-12 in a concentration-dependent manner; however, caspase activation was significantly reduced (P < 0.05) with VRP-034. This study demonstrates that VRP-034 significantly reduces nephrotoxicity compared to marketed PMB within a 3D microphysiological system, suggesting its potential to enable the use of full therapeutic doses of PMB with an improved safety profile, addressing the need for less nephrotoxic polymyxin antibiotics.


Sujet(s)
Cystatine C , Tubules contournés proximaux , Polymyxine B , Polymyxine B/pharmacologie , Humains , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Récepteur cellulaire-1 du virus de l'hépatite A/métabolisme , Cytochromes c/métabolisme , Antibactériens/pharmacologie , Laboratoires sur puces , Survie cellulaire/effets des médicaments et des substances chimiques , Marqueurs biologiques/métabolisme , Interleukine-6/métabolisme , Caspase-3/métabolisme , Lignée cellulaire , Caspase-9/métabolisme , Interleukine-8/métabolisme , Caspase 8/métabolisme , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
10.
Am J Physiol Cell Physiol ; 327(4): C1094-C1110, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39250817

RÉSUMÉ

The class 3 phosphatidylinositol 3-kinase (Pik3c3) plays critical roles in regulating autophagy, endocytosis, and nutrient sensing, but its expression profile in the kidney remains undefined. Recently, we validated a Pik3c3 antibody through immunofluorescence staining of kidney tissues from cell type-specific Pik3c3 knockout mice. Immunohistochemistry unveiled significant disparities in Pik3c3 expression levels across various kidney cell types. Notably, renal interstitial cells exhibit minimal Pik3c3 expression. Further, coimmunofluorescence staining, utilizing nephron segment- or cell type-specific markers, revealed nearly undetectable levels of Pik3c3 expression in glomerular mesangial cells and endothelial cells. Intriguingly, although podocytes exhibit the highest Pik3c3 expression levels among all kidney cell types, the renal proximal tubule cells (RPTCs) express the highest level of Pik3c3 among all renal tubules. RPTCs are known to express the highest level of the epidermal growth factor receptor (EGFR) in adult kidneys; however, the role of Pik3c3 in EGFR signaling within RPTCs remains unexplored. Therefore, we conducted additional cell culture studies. The results demonstrated that Pik3c3 inhibition significantly delayed EGF-stimulated EGFR degradation and the termination of EGFR signaling in RPTCs. Mechanistically, Pik3c3 inhibition surprisingly did not affect the initial endocytosis process but instead impeded the lysosomal degradation of EGFR. In summary, this study defines, for the first time, the expression profile of Pik3c3 in the mouse kidney and also highlights a pivotal role of Pik3c3 in the proximal tubule cells. These findings shed light on the intricate mechanisms underlying Pik3c3-mediated regulation of EGFR signaling, providing valuable insights into the role of Pik3c3 in renal cell physiology. NEW & NOTEWORTHY This is the first report defining the class 3 phosphatidylinositol 3-kinase (Pik3c3) expression profile in the kidney. Pik3c3 is nearly absent in renal interstitial cells, glomerular mesangial cells, and endothelial cells. Remarkably, glomerular podocytes express the highest Pik3c3 level in the kidney. However, the proximal tubule exhibits the highest expression level among all renal tubules. This study also unveils the pivotal role of Pik3c3 in regulating EGFR degradation and signaling termination in RPTCs, furthering our understanding of Pik3c3 in renal cell physiology.


Sujet(s)
Phosphatidylinositol 3-kinases de classe III , Récepteurs ErbB , Tubules contournés proximaux , Souris knockout , Animaux , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/enzymologie , Tubules contournés proximaux/cytologie , Souris , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Phosphatidylinositol 3-kinases de classe III/métabolisme , Phosphatidylinositol 3-kinases de classe III/génétique , Transduction du signal , Souris de lignée C57BL , Mâle , Analyse de profil d'expression de gènes/méthodes , Podocytes/métabolisme , Podocytes/enzymologie
11.
Kidney Int ; 106(4): 560-562, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39304269

RÉSUMÉ

Acute kidney injury is a devasting clinical syndrome resulting from multiple causes, characterized by an abrupt deterioration of kidney function for which there is no pharmacologic treatment. Cilastatin has demonstrated direct nephroprotective effects in acute kidney injury and now is shown to be effective to specifically target therapeutically loaded nanoparticles to the proximal tubule to treat acute kidney injury.


Sujet(s)
Atteinte rénale aigüe , Cilastatine , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/prévention et contrôle , Humains , Cilastatine/usage thérapeutique , Cilastatine/administration et posologie , Animaux , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme , Nanoparticules
12.
In Vivo ; 38(5): 2107-2114, 2024.
Article de Anglais | MEDLINE | ID: mdl-39187331

RÉSUMÉ

BACKGROUND/AIM: Angiotensinogen (AGT), a precursor of angiotensin II (AngII), contributes to regulating (patho)physiological conditions, including blood pressure changes, inflammation, and kidney fibrosis. However, the precise role of tissue-specific AGT in kidney fibrosis independent of blood pressure remains to be fully understood. This study investigated the source of intrarenal AGT and its role in kidney injury and fibrosis during obstructive nephropathy. MATERIALS AND METHODS: Proximal tubule- (PT, major source secreting AGT in the kidney; PKO) or liver- (major source of circulating AGT; LKO) AGT knockout (KO) mice were subjected to unilateral ureteral obstruction (UUO), a blood pressure-independent fibrosis model. RESULTS: UUO increased AGT mRNA and protein levels in the kidneys. PKO decreased AGT mRNA, but LKO enhanced it in UUO kidneys compared with the control. In contrast, the intrarenal protein levels of AGT increased in PKO, but not in LKO in UUO kidneys, indicating that the liver is a major source of intrarenal AGT protein. Expression of megalin, a PT receptor involved in the uptake of circulating AGT, was down-regulated in UUO kidneys and was independent of PKO or LKO. However, none of these changes prevented UUO-induced tubular injury and kidney fibrosis. CONCLUSION: Hepatic and proximal tubule AGT play distinct roles in contributing to intrarenal AGT levels during UUO, and their genetic inhibitions fail to prevent kidney injury and fibrosis, suggesting a highly complicated signaling pathway of the renin-angiotensin system and an associated compensatory mechanism in obstructive nephropathy.


Sujet(s)
Angiotensinogène , Modèles animaux de maladie humaine , Fibrose , Rein , Souris knockout , Obstruction urétérale , Animaux , Souris , Angiotensinogène/métabolisme , Angiotensinogène/génétique , Rein/métabolisme , Rein/anatomopathologie , Maladies du rein/métabolisme , Maladies du rein/étiologie , Maladies du rein/anatomopathologie , Maladies du rein/génétique , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Protéine-2 apparentée au récepteur des LDL/métabolisme , Protéine-2 apparentée au récepteur des LDL/génétique , Obstruction urétérale/métabolisme , Obstruction urétérale/complications , Obstruction urétérale/génétique , Obstruction urétérale/anatomopathologie
13.
Am J Physiol Renal Physiol ; 327(4): F610-F622, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39116349

RÉSUMÉ

Studies in animal models have suggested a linkage between the inflammatory response to injury and subsequent nephron loss during the acute kidney injury (AKI) to chronic kidney disease (CKD) transition. Failure of normal repair during the CKD transition correlates with de novo expression of vascular cell adhesion protein-1 (VCAM-1) by a subset of injured proximal tubule cells. This study identified the role of VCAM-1 expression in promoting the failed repair state. Single-cell transcriptome analysis of patients with AKI and CKD and whole kidney RNA and protein analyses of mouse models of CKD confirmed a marked increase of VCAM-1 expression in the proximal tubules of injured kidneys. In immortalized mouse proximal tubular cells and primary cultured renal cells (PCRCs), VCAM-1 expression was induced by proinflammatory cytokines including tumor necrosis factor (TNF)-α and interleukin (IL)-1ß. Analyses of bulk RNA sequencing of TNF-α-treated primary cultured renal cells or pseudo-bulk RNA sequencing of biopsies from Kidney Precision Medicine Project datasets indicated activation of NF-κB and an enrichment of inflammatory response and cell adhesion pathways in VCAM-1-positive cells. Pharmacological inhibition of NF-κB signaling or genetic deletion of myeloid differentiation factor 88 and TIR domain-containing adapter-inducing interferon-ß suppressed TNF-α- and IL-1ß-induced VCAM-1 expression in vitro. TNF-α stimulation or overexpression of VCAM-1 significantly increased splenocyte adhesion to the mouse proximal tubular monolayer in culture. These results demonstrate that persistence of proinflammatory cytokines after AKI can induce NF-κB-dependent VCAM-1 expression by proximal tubule cells, mediating increased immune cell adhesion to the tubule and thus promoting further tubule injury and greater risk of progression from AKI to CKD.NEW & NOTEWORTHY We demonstrated the induction of VCAM-1 and its biological function in proximal tubules. We found that proinflammatory cytokines (TNF-α and IL-1ß) significantly induced VCAM-1 expression via NF-κB signaling pathway. TNF-α treatment or overexpression of VCAM-1 in immortalized MPT cells increased CD45+ splenocyte adhesion. Pharmacological inhibition of NF-κB or genetic deletion of Vcam1 suppressed TNF-α-induced splenocyte adhesion in vitro, suggesting that VCAM-1 mediates proximal tubular-immune cell cross talk in failed tubule recovery during AKI-to-CKD transition.


Sujet(s)
Atteinte rénale aigüe , Tubules contournés proximaux , Insuffisance rénale chronique , Molécule-1 d'adhérence des cellules vasculaires , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Molécule-1 d'adhérence des cellules vasculaires/génétique , Animaux , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/immunologie , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/immunologie , Atteinte rénale aigüe/génétique , Humains , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/immunologie , Insuffisance rénale chronique/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Transduction du signal , Souris de lignée C57BL , Facteur de transcription NF-kappa B/métabolisme , Souris , Évolution de la maladie , Adhérence cellulaire/effets des médicaments et des substances chimiques
14.
Eur J Pharmacol ; 982: 176931, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39182553

RÉSUMÉ

Renal fibrosis is among the major factors contributing to the development of chronic kidney disease. In this regard, although N6-methyladenosine (m6A) modification and micro-RNAs (miRNAs) have been established to play key roles in diverse physiological processes and disease/disorder development, further research is required to identify the probable mechanisms and processes associated with their involvement in renal fibrosis. In this study, we show that transforming growth factor ß1 (TGF-ß1)-induced human proximal tubule epithelial cells (HK2) are characterized by dose-dependently higher methyltransferase-like 3 (METTL3) expression. Furthermore, METTL3 was found to enhance pri-miR-199a-3p maturation and miR-199a-3p expression in an m6A-dependent manner, whereas miR-199a-3p sponges prostate apoptotic response 4 (Par4), thereby regulating its expression. Collectively, our findings in this study indicate that the METTL3/miR-199a-3p/Par4 axis plays a key role in the development of obstructive nephrogenic fibrosis.


Sujet(s)
Fibrose , Methyltransferases , microARN , Facteur de croissance transformant bêta-1 , Animaux , Humains , Adénosine/analogues et dérivés , Adénosine/métabolisme , Lignée cellulaire , Régulation de l'expression des gènes , Maladies du rein/génétique , Maladies du rein/anatomopathologie , Maladies du rein/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme , Methyltransferases/génétique , Methyltransferases/métabolisme , microARN/génétique , microARN/métabolisme , Transduction du signal/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Mâle , Souris
16.
Mol Pharm ; 21(9): 4603-4617, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39166754

RÉSUMÉ

Modulation of the transport-mediated active uptake by human serum albumin (HSA) for highly protein-bound substrates has been reported and improved the in vitro-to-in vivo extrapolation (IVIVE) of hepatic clearance. However, evidence for the relevance of such a phenomenon in the case of renal transporters is sparse. In this study, transport of renal organic anion transporter 1 or 3 (OAT1/3) substrates into conditionally immortalized proximal tubular epithelial cells transduced with OAT1/3 was measured in the presence and absence of 1 and 4% HSA while keeping the unbound substrate concentration constant (based on measured fraction unbound, fu,inc). In the presence of 4% HSA, the unbound intrinsic active uptake clearance (CLint,u,active) of six highly protein-bound substrates increased substantially relative to the HSA-free control (3.5- to 122-fold for the OAT1 CLint,u,active, and up to 28-fold for the OAT3 CLint,u,active). The albumin-mediated uptake effect (fold increase in CLint,u,active) was more pronounced with highly bound substrates compared to no effect seen for weakly protein-bound substrates adefovir (OAT1-specific) and oseltamivir carboxylate (OAT3-specific). The relationship between OAT1/3 CLint,u,active and fu,inc agreed with the facilitated-dissociation model; a relationship was established between the albumin-mediated fold change in CLint,u,active and fu,inc for both the OAT1 and OAT3, with implications for IVIVE modeling. The relative activity factor and the relative expression factor based on global proteomic quantification of in vitro OAT1/3 expression were applied for IVIVE of renal clearance. The inclusion of HSA improved the bottom-up prediction of the level of OAT1/3-mediated secretion and renal clearance (CLsec and CLr), in contrast to the underprediction observed with the control (HSA-free) scenario. For the first time, this study confirmed the presence of the albumin-mediated uptake effect with renal OAT1/3 transporters; the extent of the effect was more pronounced for highly protein-bound substrates. We recommend the inclusion of HSA in routine in vitro OAT1/3 assays due to considerable improvements in the IVIVE of CLsec and CLr.


Sujet(s)
Protéine-1 de transport d'anions organiques , Transporteurs d'anions organiques sodium-indépendants , Protéine-1 de transport d'anions organiques/métabolisme , Humains , Transporteurs d'anions organiques sodium-indépendants/métabolisme , Transport biologique/physiologie , Rein/métabolisme , Animaux , Tubules contournés proximaux/métabolisme , Sérumalbumine/métabolisme , Sérum-albumine humaine/métabolisme , Lignée cellulaire
17.
Exp Cell Res ; 442(1): 114186, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39098465

RÉSUMÉ

TGFß1 is a powerful regulator of fibrosis; secreted in a latent form, it becomes active after release from the latent complex. During tissue fibrosis, the EDA + isoform of cellular fibronectin is overexpressed. In pulmonary fibrosis it has been proposed that the fibronectin splice variant including an EDA domain (FN EDA+) activates latent TGFß. Our work investigates the potential of blocking the 'splicing in' of EDA with antisense oligonucleotides to inhibit TGFß1-induced EDA + fibronectin and to prevent the cascade of events initiated by TGFß1 in human renal proximal tubule cells (PTEC). Human primary PTEC were treated with TGFß1 for 48 h, medium removed and the cells transfected with RNase H-independent antisense oligonucleotides (ASO) designed to block EDA exon inclusion (ASO5). The efficacy of ASO to block EDA exon inclusion was assessed by EDA + fibronectin RNA and protein expression; the expression of TGFß, αSMA (α smooth muscle actin), MMP2 (matrix metalloproteinse-2), MMP9 (matrix metalloproteinse-9), Collagen I, K Cadherin and connexin 43 was analysed. Targeting antisense oligonucleotides designed to block EDA exon inclusion in fibronectin pre mRNA were effective in reducing the amount of TGFß1 -induced cellular EDA + fibronectin RNA and secreted EDA + fibronectin protein (assessed by western immunoblotting and immunocytochemistry) in human proximal tubule cells in an in vitro cell culture model. The effect was selective for EDA + exon with no effect on EDB + fibronectin RNA and total fibronectin mRNA. Exogenous TGFß1 induced endogenous TGFß, αSMA, MMP2, MMP9 and Col I mRNA. TGFß1 treatment for 48h reduced the expression of K-Cadherin and increased the expression of connexin-43. These TGFß1-induced pro-fibrotic changes were attenuated by ASO5 treatment. 48 h after the removal of exogenous TGFß, further increases in αSMA, MMP2, MMP9 was observed; ASO5 significantly inhibited this subsequent increase. ASO5 treatment also significantly inhibited ability of the cell culture medium harvested at the end of the experiment (96h) to stimulate SMAD3 reporter cells. The role of endogenous TGFß1 was confirmed by the use of a TGFß receptor inhibitor. Our results demonstrate a critical role of FN EDA+ in a cycle of TGFß driven pro-fibrotic responses in human PTEC and blocking its production with ASO technology offers a potential therapy to interrupt this vicious circle and hence limit the progression of renal fibrosis.


Sujet(s)
Épissage alternatif , Cellules épithéliales , Fibronectines , Fibrose , Tubules contournés proximaux , Oligonucléotides antisens , Facteur de croissance transformant bêta-1 , Humains , Fibronectines/métabolisme , Fibronectines/génétique , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/cytologie , Oligonucléotides antisens/pharmacologie , Oligonucléotides antisens/génétique , Fibrose/métabolisme , Épissage alternatif/génétique , Facteur de croissance transformant bêta-1/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules cultivées , Communication autocrine , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 2/génétique , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique
18.
Am J Physiol Endocrinol Metab ; 327(4): E512-E523, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39140974

RÉSUMÉ

Diabetic kidney disease (DKD) remains as one of the leading long-term complications of type 2 diabetic mellitus (T2DM). Studies have shown that decreased expression of glucose-6-phosphate dehydrogenase (G6PD) plays an important role in DKD. However, the upstream and downstream pathways of G6PD downregulation leading to DKD have not been elucidated. We conducted a series of studies including clinical study, animal studies, and in vitro studies to explore this. First, a total of 90 subjects were evaluated including 30 healthy subjects, 30 patients with T2DM, and 30 patients with DKD. The urinary G6PD activity and its association with the clinical markers were analyzed. Multivariate linear regression analysis was used to analyze the risk factors of urinary G6PD in these patients. Then, microRNAs that were differentially expressed in urine and could bind and degrade G6PD were screened and verified in patients with DKD. After that, high glucose (HG)-cultured human kidney cells (HK-2) and Zucker diabetic fatty (ZDF) rats were used to test the roles of miR-7977/G6PD/albumin-induced autophagy in DKD. Beclin and P62 were used as markers of kidney autophagy indicators. A dual-luciferase reporter assay system was used to test the binding of G6PD by mir-7977. The plasma and urinary G6PD activity were decreased significantly in patients with DKD, accompanied by increased urinary mir-7977 level. The fasting plasma glucose (FPG), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and urinary albumin excretion were independent predictors of urinary G6PD activity, according to multiple linear regression analysis. The increased expression of miR-7977 and decreased expression of G6PD were also found in the kidney of ZDF rats with early renal tubular damage. The correlation analysis showed that beclin protein expression levels were positively correlated with kidney G6PD activity, whereas P62 protein expression was negatively correlated with kidney G6PD activity in rats. In HK-2 cells cultured with normal situation, a low level of albumin could induce autophagy along with the stimulation of G6PD, although this was impaired under high glucose. Overexpression of G6PD reversed albumin-induced autophagy in HK-2 cells under high glucose. Further study revealed that G6PD was a downstream target of miR-7977. Inhibition of miR-7977 expression led to significantly increased expression of G6PD and reversed the effects of high glucose on albumin-induced autophagy. In conclusion, our study supports a new mechanism of G6PD downregulation in DKD. Therapeutic measures targeting the miR-7977/G6PD/autophagy signaling pathway may help in the prevention and treatment of DKD.NEW & NOTEWORTHY This study provides new evidence that reduced glucose-6-phosphate dehydrogenase (G6PD) may damage the endocytosis of renal tubular epithelial cells by reducing albumin-induced autophagy. More importantly, for the first time, our study has provided evidence from humans that the decrease in urinary G6PD activity is positively associated with renal injury, and abnormal glucose and lipid metabolism may be important reasons for reduced G6PD levels. Increased miR-7977 may at least in part explain the downregulation of G6PD.


Sujet(s)
Autophagie , Diabète de type 2 , Néphropathies diabétiques , Glucose 6-phosphate dehydrogenase , microARN , Humains , microARN/génétique , microARN/métabolisme , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/génétique , Néphropathies diabétiques/urine , Autophagie/physiologie , Mâle , Animaux , Adulte d'âge moyen , Rats , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Femelle , Diabète de type 2/métabolisme , Diabète de type 2/complications , Diabète de type 2/génétique , Diabète de type 2/urine , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Rat Zucker , Études cas-témoins , Cellules épithéliales/métabolisme , Adulte , Lignée cellulaire , Sujet âgé
19.
FASEB J ; 38(16): e23884, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39135512

RÉSUMÉ

The inhibition of the autophagolysosomal pathway mediated by transcription factor EB (TFEB) inactivation in proximal tubular epithelial cells (TECs) is a key mechanism of TEC injury in diabetic kidney disease (DKD). Acetylation is a novel mechanism that regulates TFEB activity. However, there are currently no studies on whether the adjustment of the acetylation level of TFEB can reduce the damage of diabetic TECs. In this study, we investigated the effect of Trichostatin A (TSA), a typical deacetylase inhibitor, on TFEB activity and damage to TECs in both in vivo and in vitro models of DKD. Here, we show that TSA treatment can alleviate the pathological damage of glomeruli and renal tubules and delay the DKD progression in db/db mice, which is associated with the increased expression of TFEB and its downstream genes. In vitro studies further confirmed that TSA treatment can upregulate the acetylation level of TFEB, promote its nuclear translocation, and activate the expression of its downstream genes, thereby reducing the apoptosis level of TECs. TFEB deletion or HDAC6 knockdown in TECs can counteract the activation effect of TSA on autophagolysosomal pathway. We also found that TFEB enhances the transcription of Tfeb through binding to its promoter and promotes its own expression. Our results, thus, provide a novel therapeutic mechanism for DKD that the alleviation of TEC damage by activating the autophagic lysosomal pathway through upregulating TFEB acetylation can, thus, delay DKD progression.


Sujet(s)
Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Néphropathies diabétiques , Cellules épithéliales , Inhibiteurs de désacétylase d'histone , Acides hydroxamiques , Tubules contournés proximaux , Animaux , 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 , Néphropathies diabétiques/métabolisme , Souris , Acétylation , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Acides hydroxamiques/pharmacologie , Inhibiteurs de désacétylase d'histone/pharmacologie , Mâle , Souris de lignée C57BL , Autophagie/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
20.
Sci Rep ; 14(1): 19443, 2024 08 21.
Article de Anglais | MEDLINE | ID: mdl-39169052

RÉSUMÉ

Cisplatin-induced nephrotoxicity restricts its clinical use against solid tumors. The present study elucidated the pharmacological effects of Renogrit, a plant-derived prescription medicine, using cisplatin-induced human renal proximal tubular (HK-2) cells and Caenorhabditis elegans. Quantification of phytochemicals in Renogrit was performed on HPTLC and UHPLC platforms. Renogrit was assessed in vitro in HK-2 cells post-exposure to clinically relevant concentration of cisplatin. It was observed that renoprotective properties of Renogrit against cisplatin-induced injury stem from its ability to regulate renal injury markers (KIM-1, NAG levels; NGAL mRNA expression), redox imbalance (ROS generation; GST levels), and mitochondrial dysfunction (mitochondrial membrane potential; SKN-1, HSP-60 expression). Renogrit was also found to modulate apoptosis (EGL-1 mRNA expression; protein levels of p-ERK, p-JNK, p-p38, c-PARP1), necroptosis (intracellular calcium accumulation; RIPK1, RIPK3, MLKL mRNA expression), mitophagy (lysosome population; mRNA expression of PINK1, PDR1; protein levels of p-PINK1, LC3B), and inflammation (IL-1ß activity; protein levels of LXR-α). More importantly, Renogrit treatment did not hamper normal anti-proliferative effects of cisplatin as observed from cytotoxicity analysis on MCF-7, A549, SiHa, and T24 human cancer cells. Taken together, Renogrit could be a potential clinical candidate to mitigate cisplatin-induced nephrotoxicity without compromising the anti-neoplastic properties of cisplatin.


Sujet(s)
Apoptose , Caenorhabditis elegans , Cisplatine , Mitophagie , Cisplatine/effets indésirables , Cisplatine/toxicité , Animaux , Humains , Mitophagie/effets des médicaments et des substances chimiques , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Extraits de plantes/pharmacologie , Tubules rénaux/effets des médicaments et des substances chimiques , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/toxicité , Antinéoplasiques/effets indésirables , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE