Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.136
Filtrer
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Sci Total Environ ; 949: 175159, 2024 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-39094650

RÉSUMÉ

Bisphenol P (BPP) has been detected in human biological samples; however studies on its nephrotoxicity are scarce. Given the susceptibility of kidneys to endocrine-disrupting chemicals, there is an urgent need to investigate the renal toxicity of BPP. This study aimed to evaluate the effects of different concentrations of BPPs on the kidneys of C57BL/6 mice and elucidate the underlying mechanisms of renal damage using a combination of mouse renal transcriptomic data and human renal proximal tubular epithelial cells (HK-2). Mice were exposed to BPP (0, 0.3, 30, 3000 µg/kg bw/d) via gavage for 5 weeks. Renal injury was assessed based on changes in body and kidney weights, serum renal function indices, and histopathological examination. Transcriptomic analysis identified differentially expressed genes and pathways, whereas cellular assays were used to measure cell viability, reactive oxygen species (ROS), apoptosis, and the expression of key genes and proteins. The results show that BPP exposure induces renal injury, as evidenced by increased body weight, abnormal renal function indices, and renal tissue damage. Transcriptomic analysis revealed alterations in genes and pathways related to oxidative stress, p53 signaling, autophagy, and apoptosis. Cellular experiments confirmed that BPP induces oxidative stress and apoptosis. Furthermore, BPP exposure significantly inhibits autophagy, potentially exacerbating apoptosis and contributing to kidney injury. Treatment with a ROS inhibitor (N-Acetylcysteine, NAC) mitigated BPP-induced autophagy inhibition and apoptosis, implicating oxidative stress as a key factor. BPP exposure may lead to renal injury through excessive ROS accumulation, oxidative stress, inflammatory responses, autophagy inhibition, and increased apoptosis. The effects of NAC highlight the role of oxidative stress in BPP-induced nephrotoxicity. These findings enhance our understanding of BPP-induced nephrotoxicity and underscore the need to control BPP exposure to prevent renal disease. This study emphasized the importance of evaluating the safety of new Bisphenol A analogs, including BPP, in environmental toxicology.


Sujet(s)
Cellules épithéliales , Souris de lignée C57BL , Stress oxydatif , Phénols , Animaux , Humains , Souris , Apoptose/effets des médicaments et des substances chimiques , Composés benzhydryliques/toxicité , Perturbateurs endocriniens/toxicité , Cellules épithéliales/effets des médicaments et des substances chimiques , Rein/cytologie , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Phénols/toxicité , Espèces réactives de l'oxygène/métabolisme
7.
J Biochem Mol Toxicol ; 38(9): e23777, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39165170

RÉSUMÉ

Tramadol (TR), a commonly prescribed pain reliever for moderate to severe pain, has been associated with kidney damage. This study investigates TR-induced nephrotoxicity mechanisms, focusing on its effects on renal proximal tubular cells (PTCs). The study findings demonstrate that TR disrupts PTC bioenergetic processes, leading to oxidative stress and inflammation. Significant toxicity to PTCs was observed with estimated effective concentration 50 values of 9.8 and 11.5 µM based on 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase assays, respectively. TR also interferes with the function of PTC transporters, including organic cation uptake transporter 1, organic cation transporter 2, P-glycoprotein, and multidrug resistance protein 2. Furthermore, bioenergetics assays showed that TR reduced the activities of mitochondrial complexes I and III, adenosine triphosphate production, mitochondrial membrane potential, and oxygen consumption rate while increasing lactate release. TR also increased the production of reactive oxygen species, lipid peroxidation thiobarbituric acid reactive substances end products, and the expression of the NRf2 gene while decreasing reduced glutathione (GSH-R) stores and catalase and superoxide dismutase antioxidant activities. Additionally, TR increased the production of inflammatory cytokines (TNF-α and IL-6) and their coding genes expression. Interestingly, the study found that antioxidants like GSH-R, inhibitors of IL-6 and TNF-α, and mitochondrial activating Co-Q10 could protect cells against TR-induced cytotoxicity. The study suggests that TR causes nephrotoxicity by disrupting bioenergetic processes, causing oxidative stress and inflammation, but antioxidants and agents targeting mitochondria may have protective and curative potential.


Sujet(s)
Métabolisme énergétique , Inflammation , Stress oxydatif , Tramadol , Stress oxydatif/effets des médicaments et des substances chimiques , Inflammation/induit chimiquement , Inflammation/métabolisme , Tramadol/effets indésirables , Tramadol/pharmacologie , Métabolisme énergétique/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 , Humains , Lignée cellulaire , Animaux , Analgésiques morphiniques/effets indésirables , Analgésiques morphiniques/pharmacologie
8.
Cell Signal ; 122: 111308, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39059756

RÉSUMÉ

BACKGROUND: The protection of the diabetic kidney by Empagliflozin (EMPA) is attributed to its interaction with the sodium glucose cotransporter 2 located on proximal tubular epithelial cells (PTECs). Estrogen-related receptor α (ESRRA), known for its high expression in PTECs and association with mitochondrial biogenesis, plays a crucial role in this process. This study aimed to explore the impact of ESRRA on mitochondrial mass in diabetic tubular injury and elucidate the mechanism underlying the protective effects of EMPA. METHODS: Mitochondrial changes in PTECs of 16-week-old diabetic mice were assessed using transmission electron microscopy (TEM) and RNA-sequences. In vivo, EMPA administration was carried out in db/db mice for 8 weeks, while in vitro experiments involved modifying ESRRA expression in HK2 cells using pcDNA-ESRRA or EMPA. RESULTS: Evaluation through TEM revealed reduced mitochondrial mass and swollen mitochondria in PTECs, whereas no significant changes were observed under light microscopy. Analysis of RNA-sequences identified 110 downregulated genes, including Esrra, associated with mitochondrial function. Notably, ESRRA overexpression rescued the loss of mitochondrial mass induced by high glucose (HG) in HK2 cells. EMPA treatment ameliorated the ultrastructural alterations and mitigated the downregulation of ESRRA both in db/db mice and HG-treated HK2 cells. CONCLUSION: The diminished expression of ESRRA is implicated in the decline of mitochondrial mass in PTECs during the early stages of diabetes, highlighting it as a key target of EMPA for preventing the progression of diabetic kidney injury.


Sujet(s)
Composés benzhydryliques , Diabète expérimental , Néphropathies diabétiques , Glucosides , Mitochondries , Animaux , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/ultrastructure , Composés benzhydryliques/pharmacologie , Souris , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Diabète expérimental/traitement médicamenteux , Diabète expérimental/anatomopathologie , Diabète expérimental/métabolisme , Mâle , ERRalpha Estrogen-Related Receptor , Humains , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/ultrastructure , Souris de lignée C57BL , Lignée cellulaire , Récepteurs des oestrogènes/métabolisme
9.
Chem Biol Interact ; 399: 111123, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-38964638

RÉSUMÉ

Pharmacokinetic changes induced by radiation following radiotherapy ("RT-PK" phenomenon) are of great significance to the effectiveness and safety of chemotherapeutic agents in clinical settings. The aims of this study were to clarify the organic anion transporters (Oats) involved in the "RT-PK" phenomenon of bestatin in rats following X-ray irradiation and to elucidate its potential mechanism via vitamin D signalling. Pharmacokinetic studies, uptake assays using rat kidney slices and primary proximal tubule cells, and molecular biological studies were performed. Significantly increased plasma concentrations and systemic exposure to bestatin were observed at 24 and 48 h following abdominal X-ray irradiation, regardless of oral or intravenous administration of the drugs in rats. Reduced renal clearance and cumulative urinary excretion of bestatin were observed at 24 and 48 h post-irradiation in rats following intravenous administration. The uptake of the probe substrates p-aminohippuric acid and oestrone 3-sulfate sodium in vitro and the expression of Oat1 and Oat3 in vivo were reduced in the corresponding models following irradiation. Moreover, the upregulation of the vitamin D receptor (Vdr) in mRNA and protein levels negatively correlated with the expressions and functions of Oat1 and Oat3 following irradiation. Additionally, elevated plasma urea nitrogen levels and histopathological changes were observed in rats after exposure to irradiation. The "RT-PK" phenomenon of bestatin occurs in rats after exposure to irradiation, possibly resulting in the regulation of the expressions and activities of renal Oats via activation of the Vdr signalling pathway.


Sujet(s)
Régulation négative , Rein , Récepteur calcitriol , Animaux , Rats , Récepteur calcitriol/métabolisme , Mâle , Rein/métabolisme , Rein/effets des radiations , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/effets des radiations , Rat Sprague-Dawley , Rayons X , Protéine-1 de transport d'anions organiques/métabolisme , Protéine-1 de transport d'anions organiques/génétique , Transporteurs d'anions organiques/métabolisme , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/effets des radiations , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Leucine/analogues et dérivés
10.
Kidney Int ; 106(4): 597-610, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39067856

RÉSUMÉ

Acute kidney injury (AKI) increases the risk of in-hospital death, adds to expense of care, and risk of early chronic kidney disease. AKI often follows an acute event such that timely treatment could ameliorate AKI and potentially reduce the risk of additional disease. Despite therapeutic success of dexamethasone in animal models, clinical trials have not demonstrated broad success. To improve the safety and efficacy of dexamethasone for AKI, we developed and characterized a novel, kidney-specific nanoparticle enabling specific within-kidney targeting to proximal tubular epithelial cells provided by the megalin ligand cilastatin. Cilastatin and dexamethasone were complexed to H-Dot nanoparticles, which were constructed from generally recognized as safe components. Cilastatin/Dexamethasone/H-Dot nanotherapeutics were found to be stable at plasma pH and demonstrated salutary release kinetics at urine pH. In vivo, they were specifically biodistributed to the kidney and bladder, with 75% recovery in the urine and with reduced systemic toxicity compared to native dexamethasone. Cilastatin complexation conferred proximal tubular epithelial cell specificity within the kidney in vivo and enabled dexamethasone delivery to the proximal tubular epithelial cell nucleus in vitro. The Cilastatin/Dexamethasone/H-Dot nanotherapeutic improved kidney function and reduced kidney cellular injury when administered to male C57BL/6 mice in two translational models of AKI (rhabdomyolysis and bilateral ischemia reperfusion). Thus, our design-based targeting and therapeutic loading of a kidney-specific nanoparticle resulted in preservation of the efficacy of dexamethasone, combined with reduced off-target disposition and toxic effects. Hence, our study illustrates a potential strategy to target AKI and other diseases of the kidney.


Sujet(s)
Atteinte rénale aigüe , Dexaméthasone , Cellules épithéliales , Tubules contournés proximaux , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/prévention et contrôle , Animaux , Dexaméthasone/pharmacologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme , Humains , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Nanoparticules , Mâle , Modèles animaux de maladie humaine , Souris , Souris de lignée C57BL , Protéine-2 apparentée au récepteur des LDL/métabolisme
11.
Biochim Biophys Acta Gen Subj ; 1868(10): 130684, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39084330

RÉSUMÉ

It is well-established that dysfunction of megalin-mediated albumin endocytosis by proximal tubule epithelial cells (PTECs) and the activation of the Renin-Angiotensin System (RAS) play significant roles in the development of Diabetic Kidney Disease (DKD). However, the precise correlation between these factors still requires further investigation. In this study, we aimed to elucidate the potential role of angiotensin II (Ang II), a known effector of RAS, as the mediator of albumin endocytosis dysfunction induced by high glucose (HG) in PTECs. To achieve this, we utilized LLC-PK1 and HK-2 cells, which are well-established in vitro models of PTECs. Using albumin-FITC or DQ-albumin as tracers, we observed that incubation of LLC-PK1 and HK-2 cells with HG (25 mM for 48 h) significantly reduced canonical receptor-mediated albumin endocytosis, primarily due to the decrease in megalin expression. HG increased the concentration of Ang II in the LLC-PK1 cell supernatant, a phenomenon associated with an increase in angiotensin-converting enzyme (ACE) expression and a decrease in prolyl carboxypeptidase (PRCP) expression. ACE type 2 (ACE2) expression remained unchanged. To investigate the potential impact of Ang II on HG effects, the cells were co-incubated with angiotensin receptor inhibitors. Only co-incubation with 10-7 M losartan (an antagonist for type 1 angiotensin receptor, AT1R) attenuated the inhibitory effect of HG on albumin endocytosis, as well as megalin expression. Our findings contribute to understanding the genesis of tubular albuminuria observed in the early stages of DKD, which involves the activation of the Ang II/AT1R axis by HG.


Sujet(s)
Albumines , Angiotensine-II , Endocytose , Cellules épithéliales , Glucose , Tubules contournés proximaux , Récepteur de type 1 à l'angiotensine-II , Endocytose/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Angiotensine-II/pharmacologie , Glucose/métabolisme , Glucose/pharmacologie , Récepteur de type 1 à l'angiotensine-II/métabolisme , Animaux , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Humains , Albumines/métabolisme , Suidae , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Protéine-2 apparentée au récepteur des LDL/métabolisme , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire , Losartan/pharmacologie
12.
Toxicol Mech Methods ; 34(9): 1022-1034, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39034811

RÉSUMÉ

The harmful effects of PM2.5 on human health, including an increased risk of chronic kidney disease (CKD), have raised a lot of attention, but the underlying mechanisms are unclear. We used the Shanghai Meteorological and Environmental Animal Exposure System (Shanghai-METAS) to simulate the inhalation of PM2.5 in the real environment and established an animal model by exposing C57BL/6 mice to filtered air (FA) and Particulate Matter (PM2.5) for 8 weeks. PM2.5 impaired the renal function of the mice, and the renal tubules underwent destructive changes. Analysis of NHANES data showed a correlation between reduced kidney function and higher blood levels of PM2.5 components, polychlorinated biphenyls (PCBs) and dioxins, which are Aryl hydrocarbon Receptor (AhR) ligands. PM2.5 exposure induced higher levels of AhR and CYP1A1 and oxidative stress as evidenced by the higher levels of ROS, MDA, and GSSG in kidneys of mice. PM2.5 exposure led to AhR overexpression and nuclear translocation in proximal renal tubular epithelial cells. Inhibition of AhR reduced CYP1A1 expression and PM2.5-increased levels of ROS, MDA and GSSG. Our study suggested metformin can mitigate PM2.5-induced oxidative stress by inhibiting the AhR/CYP1A1 pathway. These findings illuminated the role of AhR/CYP1A1 pathway in PM2.5-induced kidney injury and the protective effect of metformin on PM2.5-induced cellular damage, offering new insights for air pollution-related renal diseases.


Sujet(s)
Cytochrome P-450 CYP1A1 , Cellules épithéliales , Tubules contournés proximaux , Metformine , Souris de lignée C57BL , Stress oxydatif , Matière particulaire , Récepteurs à hydrocarbure aromatique , Transduction du signal , Animaux , Récepteurs à hydrocarbure aromatique/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Matière particulaire/toxicité , Cytochrome P-450 CYP1A1/métabolisme , Cytochrome P-450 CYP1A1/génétique , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/métabolisme , Metformine/pharmacologie , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Polluants atmosphériques/toxicité , Souris , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Lignée cellulaire , Humains
13.
Methods Mol Biol ; 2823: 95-108, 2024.
Article de Anglais | MEDLINE | ID: mdl-39052216

RÉSUMÉ

Three-dimensional (3D) cell culture creates a more physiologically relevant environment for enhanced drug screening capabilities using microcarriers. An automated 3D system that integrates robotic manipulators, liquid handling systems, sensors, and environment control systems has the capacity to handle multiple samples in parallel, perform repetitive tasks, and provide real-time monitoring and analysis. This chapter describes a potential 3D cell culture drug screening model by combining renal proximal tubule cells as a representative normal cell line with cancer cell lines. This combination is subjected to drug screening to evaluate the drug's efficacy in suppressing cancer cells while minimizing impact on normal cells with the added benefit of having the ability to separate the two cell types by magnetic isolation for high content screens including mass spectrometry-based proteomics. This study presents advancements in 3D cell culture techniques, emphasizing the importance of automation and the potential of microcarriers in drug screening and disease modeling.


Sujet(s)
Techniques de cultures cellulaires tridimensionnelles , Humains , Techniques de cultures cellulaires tridimensionnelles/méthodes , Lignée cellulaire tumorale , Évaluation préclinique de médicament/méthodes , Tests de criblage d'agents antitumoraux/méthodes , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Techniques de culture cellulaire/méthodes , Antinéoplasiques/pharmacologie , Automatisation , Laboratoire automatique/méthodes , Tumeurs/anatomopathologie , Tumeurs/traitement médicamenteux
14.
Sci Rep ; 14(1): 15635, 2024 07 07.
Article de Anglais | MEDLINE | ID: mdl-38972889

RÉSUMÉ

This study aimed to elucidate the influence of miR-483-3p on human renal tubular epithelial cells (HK-2) under high glucose conditions and to understand its mechanism. Human proximal tubular epithelial cells (HK-2) were exposed to 50 mmol/L glucose for 48 h to establish a renal tubular epithelial cell injury model, denoted as the high glucose group (HG group). Cells were also cultured for 48 h in a medium containing 5.5 mmol/L glucose, serving as the low glucose group. Transfection was performed in various groups: HK-2 + low glucose (control group), high glucose (50 mM) (HG group), high glucose + miR-483-3p mimics (HG + mimics group), high glucose +miR-483-3p inhibitor (HG + inhibitor group), and corresponding negative controls. Real-time quantitative polymerase chain reaction (qPCR) assessed the mRNA expression of miR-483-3p, bax, bcl-2, and caspase-3. Western blot determined the corresponding protein levels. Proliferation was assessed using the CCK-8 assay, and cell apoptosis was analyzed using the fluorescence TUNEL method. Western blot and Masson's staining were conducted to observe alterations in cell fibrosis post miR-483-3p transfection. Furthermore, a dual-luciferase assay investigated the targeting relationship between miR-483-3p and IGF-1. The CCK8 assay demonstrated that the HG + mimics group inhibited HK-2 cell proliferation, while the fluorescent TUNEL method revealed induced cell apoptosis in this group. Conversely, the HG + inhibitor group promoted cell proliferation and suppressed cell apoptosis. The HG + mimics group upregulated mRNA and protein expression of pro-apoptotic markers (bax and caspase-3), while downregulating anti-apoptotic marker (bcl-2) expression. In contrast, the HG + inhibitor group showed opposite effects. Collagen I and FN protein levels were significantly elevated in the HG + mimics group compared to controls (P < 0.05). Conversely, in the HG + inhibitor group, the protein expression of Collagen I and FN was notably reduced compared to the HG group (P < 0.05). The dual luciferase reporter assay confirmed that miR-483-3p could inhibit the luciferase activity of IGF-1's 3'-UTR region (P < 0.05). miR-483-3p exerts targeted regulation on IGF-1, promoting apoptosis and fibrosis in renal tubular epithelial cells induced by high glucose conditions.


Sujet(s)
Apoptose , Prolifération cellulaire , Cellules épithéliales , Glucose , Facteur de croissance IGF-I , Tubules rénaux , microARN , Humains , microARN/génétique , microARN/métabolisme , Glucose/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Facteur de croissance IGF-I/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Tubules rénaux/métabolisme , Tubules rénaux/cytologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Caspase-3/génétique
16.
Arch Toxicol ; 98(10): 3323-3336, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38896176

RÉSUMÉ

Ochratoxin A (OTA), a secondary fungal metabolite known for its nephrotoxic effects, is prevalent in various feeds and food items. Our recent study suggests that OTA-induced nephrotoxicity is linked to the Sigma-1 receptor (Sig-1R)-mediated mitochondrial pathway apoptosis in human proximal tubule epithelial-originated kidney-2 (HK-2) cells. However, the contribution of Sig-1R to OTA-induced nephrotoxicity involving other forms of regulated cell death, such as ferroptosis, remains unexplored. In this investigation, cell viability, malondialdehyde (MDA) levels, glutathione (GSH) levels, and protein expressions in HK-2 cells treated with OTA and/or Ferrostatin-1/blarcamesine hydrochloride/BD1063 dihydrochloride were assessed. The results indicate that a 24 h-treatment with 1 µM OTA significantly induces ferroptosis by inhibiting Sig-1R, subsequently promoting nuclear receptor coactivator 4 (NCOA4), long-chain fatty acid-CoA ligase 4 (ACSL4), arachidonate 5-lipoxygenase (ALOX5), autophagy protein 5 (ATG5), and ATG7, inhibiting ferritin heavy chain (FTH1), solute carrier family 7 member 11 (SLC7A11/xCT), glutathione peroxidase 4 (GPX4), peroxiredoxin 6 (PRDX6), and ferroptosis suppressor protein 1 (FSP1), reducing GSH levels, and increasing MDA levels (P < 0.05). In conclusion, OTA induces ferroptosis by inhibiting Sig-1R, subsequently promoting ferritinophagy, inhibiting GPX4/FSP1 antioxidant systems, reducing GSH levels, and ultimately increasing lipid peroxidation levels in vitro.


Sujet(s)
Ferroptose , Ochratoxines , Récepteur sigma , Sigma-1 Receptor , Ochratoxines/toxicité , Ferroptose/effets des médicaments et des substances chimiques , Récepteur sigma/métabolisme , Humains , Lignée cellulaire , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Tubules contournés proximaux/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Glutathion/métabolisme
17.
Am J Physiol Renal Physiol ; 327(2): F208-F223, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38870264

RÉSUMÉ

Increased dietary phosphate consumption intensifies renal phosphate burden. Several mechanisms for phosphate-induced renal tubulointerstitial fibrosis have been reported. Considering the dual nature of phosphate as both a potential renal toxin and an essential nutrient for the body, kidneys may possess inherent protective mechanisms against phosphate overload, rather than succumbing solely to injury. However, there is limited understanding of such mechanisms. To identify these mechanisms, we conducted single-cell RNA sequencing (scRNA-seq) analysis of the kidneys of control and dietary phosphate-loaded (Phos) mice at a time point when the Phos group had not yet developed tubulointerstitial fibrosis. scRNA-seq analysis identified the highest number of differentially expressed genes in the clusters belonging to proximal tubular epithelial cells (PTECs). Based on these differentially expressed genes, in silico analyses suggested that the Phos group activated peroxisome proliferator-activated receptor-α (PPAR-α) and fatty acid ß-oxidation (FAO) in the PTECs. This activation was further substantiated through various experiments, including the use of an FAO activity visualization probe. Compared with wild-type mice, Ppara knockout mice exhibited exacerbated tubulointerstitial fibrosis in response to phosphate overload. Experiments conducted with cultured PTECs demonstrated that activation of the PPAR-α/FAO pathway leads to improved cellular viability under high-phosphate conditions. The Phos group mice showed a decreased serum concentration of free fatty acids, which are endogenous PPAR-α agonists. Instead, experiments using cultured PTECs revealed that phosphate directly activates the PPAR-α/FAO pathway. These findings indicate that noncanonical metabolic reprogramming via endogenous activation of the PPAR-α/FAO pathway in PTECs is essential to counteract phosphate toxicity.NEW & NOTEWORTHY This study revealed the activation of peroxisome proliferator-activated receptor-α and fatty acid ß-oxidation in proximal tubular epithelial cells as an endogenous mechanism to protect the kidney from phosphate toxicity. These findings highlight noncanonical metabolic reprogramming as a potential target for suppressing phosphate toxicity in the kidneys.


Sujet(s)
Tubules contournés proximaux , Récepteur PPAR alpha , Phosphates , Animaux , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Phosphates/métabolisme , Phosphates/toxicité , Fibrose , Souris de lignée C57BL , Mâle , Souris , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Acides gras/métabolisme , Souris knockout , Oxydoréduction
18.
Drug Metab Dispos ; 52(9): 949-956, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-38866474

RÉSUMÉ

The role of the kidney as an excretory organ for exogenous and endogenous compounds is well recognized, but there is a wealth of data demonstrating that the kidney has significant metabolizing capacity for a variety of exogenous and endogenous compounds that in some cases surpass the liver. The induction of drug-metabolizing enzymes by some chemicals can cause drug-drug interactions and intraindividual variability in drug clearance. In this study, we evaluated the expression and induction of cytochrome P450 (P450) and UDP-glucuronosyltransferase (UGT) isoforms in 3D-cultured primary human renal proximal tubule epithelial cells (RPTEC) to elucidate their utility as models of renal drug metabolism. CYP2B6, CYP2E1, CYP3A4, CYP3A5, and all detected UGTs (UGT1A1, UGT1A4, UGT1A6, UGT1A9, and UGT2B7) mRNA levels in 3D-RPTEC were significantly higher than those in 2D-RPTEC and HK-2 cells and were close to the levels in the human kidney cortex. CYP1B1 and CYP2J2 mRNA levels in 3D-RPTEC were comparable to those in 2D-RPTEC, HK-2 cells, and the human kidney cortex. Midazolam 1'-hydroxylation, trifluoperazine N-glucuronidation, serotonin O-glucuronidation, propofol O-glucuronidation, and morphine 3-glucuronidation in the 3D-RPTEC were significantly higher than the 2D-RPTEC and comparable to those in the HepaRG cells, although bupropion, ebastine, and calcitriol hydroxylations were not different between the 2D- and 3D-RPTEC. Treatment with ligands of the aryl hydrocarbon receptor and farnesoid X receptor induced CYP1A1 and UGT2B4 expression, respectively, in 3D-RPTEC compared with 2D-RPTEC. We provided information on the expression, activity, and induction abilities of P450s and UGTs in 3D-RPTEC as an in vitro human renal metabolism model. SIGNIFICANCE STATEMENT: This study demonstrated that the expression of cytochrome P450s (P450s) and UDP-glucuronosyltransferases (UGTs) in 3D-cultured primary human renal proximal tubule epithelial cells (3D-RPTEC) was higher than those in 2D-cultured primary human renal proximal tubule epithelial cells and HK-2 cells. The results were comparable to that in the human kidney cortex. 3D-RPTEC are useful for evaluating the induction of kidney P450s, UDP-glucuronosyltransferases, and human renal drug metabolism in cellulo.


Sujet(s)
Cytochrome P-450 enzyme system , Cellules épithéliales , Glucuronosyltransferase , Tubules contournés proximaux , Humains , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Glucuronosyltransferase/métabolisme , Glucuronosyltransferase/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cytochrome P-450 enzyme system/métabolisme , Cytochrome P-450 enzyme system/génétique , Cellules cultivées , Induction enzymatique/effets des médicaments et des substances chimiques , Lignée cellulaire , Techniques de culture cellulaire/méthodes , ARN messager/métabolisme , ARN messager/génétique
19.
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
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE