Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 53
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Biochem Pharmacol ; 225: 116328, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815628

RÉSUMÉ

Early stages of diabetes are characterized by elevations of insulin and glucose concentrations. Both factors stimulate reactive oxygen species (ROS) production, leading to impairments in podocyte function and disruption of the glomerular filtration barrier. Podocytes were recently shown to be an important source of αKlotho (αKL) expression. Low blood Klotho concentrations are also associated with an increase in albuminuria, especially in patients with diabetes. We investigated whether ADAM10, which is known to cleave αKL, is activated in glomeruli and podocytes under diabetic conditions and the potential mechanisms by which ADAM10 mediates ROS production and disturbances of the glomerular filtration barrier. In cultured human podocytes, high glucose increased ADAM10 expression, shedding, and activity, NADPH oxidase activity, ROS production, and albumin permeability. These effects of glucose were inhibited when cells were pretreated with an ADAM10 inhibitor or transfected with short-hairpin ADAM10 (shADAM10) or after the addition soluble Klotho. We also observed increases in ADAM10 activity, NOX4 expression, NADPH oxidase activity, and ROS production in αKL-depleted podocytes. This was accompanied by an increase in albumin permeability in shKL-expressing podocytes. The protein expression and activity of ADAM10 also increased in isolated glomeruli and urine samples from diabetic rats. Altogether, these results reveal a new mechanism by which hyperglycemia in diabetes increases albumin permeability through ADAM10 activation and an increase in oxidative stress via NOX4 enzyme activation. Moreover, αKlotho downregulates ADAM10 activity and supports redox balance, consequently protecting the slit diaphragm of podocyteσ under hyperglycemic conditions.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Diabète expérimental , Glucuronidase , Protéines Klotho , Protéines membranaires , Podocytes , Espèces réactives de l'oxygène , Podocytes/métabolisme , Podocytes/effets des médicaments et des substances chimiques , Protéines Klotho/métabolisme , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Espèces réactives de l'oxygène/métabolisme , Humains , Animaux , Glucuronidase/métabolisme , Glucuronidase/génétique , Amyloid precursor protein secretases/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Rats , Mâle , Diabète expérimental/métabolisme , NADPH Oxidase 4/métabolisme , NADPH Oxidase 4/génétique , NADPH oxidase/métabolisme , Cellules cultivées , Glucose/métabolisme , Rat Sprague-Dawley
2.
Free Radic Biol Med ; 220: 312-323, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38740101

RÉSUMÉ

Podocytes are crucial for regulating glomerular permeability. They have foot processes that are integral to the renal filtration barrier. Understanding their energy metabolism could shed light on the pathogenesis of filtration barrier injury. Lactate has been increasingly recognized as more than a waste product and has emerged as a significant metabolic fuel and reserve. The recent identification of lactate transporters in podocytes, the expression of which is modulated by glucose levels and lactate, highlights lactate's relevance. The present study investigated the impact of lactate on podocyte respiratory efficiency and mitochondrial dynamics. We confirmed lactate oxidation in podocytes, suggesting its role in cellular energy production. Under conditions of glucose deprivation or lactate supplementation, a significant shift was seen toward oxidative phosphorylation, reflected by an increase in the oxygen consumption rate/extracellular acidification rate ratio. Notably, lactate dehydrogenase A (LDHA) and lactate dehydrogenase B (LDHB) isoforms, which are involved in lactate conversion to pyruvate, were detected in podocytes for the first time. The presence of lactate led to higher intracellular pyruvate levels, greater LDH activity, and higher LDHB expression. Furthermore, lactate exposure increased mitochondrial DNA-to-nuclear DNA ratios and resulted in upregulation of the mitochondrial biogenesis markers peroxisome proliferator-activated receptor coactivator-1α and transcription factor A mitochondrial, regardless of glucose availability. Changes in mitochondrial size and shape were observed in lactate-exposed podocytes. These findings suggest that lactate is a pivotal energy source for podocytes, especially during energy fluctuations. Understanding lactate's role in podocyte metabolism could offer insights into renal function and pathologies that involve podocyte injury.


Sujet(s)
L-Lactate dehydrogenase , Acide lactique , Dynamique mitochondriale , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Podocytes , Podocytes/métabolisme , Podocytes/anatomopathologie , Animaux , Rats , Acide lactique/métabolisme , L-Lactate dehydrogenase/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Glucose/métabolisme , Métabolisme énergétique , Lactate dehydrogenase 5/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , ADN mitochondrial/métabolisme , ADN mitochondrial/génétique , Consommation d'oxygène , Cellules cultivées , Acide pyruvique/métabolisme , Isoenzymes
3.
J Endocrinol ; 261(3)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38552310

RÉSUMÉ

Diabetic nephropathy (DN) is one of the most frequent complications of diabetes. Early stages of DN are associated with hyperinsulinemia and progressive insulin resistance in insulin-sensitive cells, including podocytes. The diabetic environment induces pathological changes, especially in podocyte bioenergetics, which is tightly linked with mitochondrial dynamics. The regulatory role of insulin in mitochondrial morphology in podocytes has not been fully elucidated. Therefore, the main goal of the present study was to investigate effects of insulin on the regulation of mitochondrial dynamics and bioenergetics in human podocytes. Biochemical analyses were performed to assess oxidative phosphorylation efficiency by measuring the oxygen consumption rate (OCR) and glycolysis by measuring the extracellular acidification rate (ECAR). mRNA and protein expression were determined by real-time polymerase chain reaction and Western blot. The intracellular mitochondrial network was visualized by MitoTracker staining. All calculations were conducted using CellProfiler software. Short-term insulin exposure exerted inhibitory effects on various parameters of oxidative respiration and adenosine triphosphate production, and glycolysis flux was elevated. After a longer time of treating cells with insulin, an increase in mitochondrial size was observed, accompanied by a reduction of expression of the mitochondrial fission markers DRP1 and FIS1 and an increase in mitophagy. Overall, we identified a previously unknown role for insulin in the regulation of oxidative respiration and glycolysis and elucidated mitochondrial dynamics in human podocytes. The present results emphasize the importance of the duration of insulin stimulation for its metabolic and molecular effects, which should be considered in clinical and experimental studies of DN.


Sujet(s)
Métabolisme énergétique , Glycolyse , Insuline , Mitochondries , Dynamique mitochondriale , Podocytes , Podocytes/métabolisme , Podocytes/effets des médicaments et des substances chimiques , Humains , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Insuline/métabolisme , Insuline/pharmacologie , Métabolisme énergétique/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Consommation d'oxygène/effets des médicaments et des substances chimiques , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Dynamines/métabolisme , Dynamines/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Phosphorylation oxydative/effets des médicaments et des substances chimiques , dGTPases/métabolisme , dGTPases/génétique , Mitophagie/effets des médicaments et des substances chimiques , Lignée cellulaire
4.
Arch Biochem Biophys ; 753: 109927, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38350532

RÉSUMÉ

Prolonged disruption in the balance of glucose can result in metabolic disorders. The kidneys play a significant role in regulating blood glucose levels. However, when exposed to chronic hyperglycemia, the kidneys' ability to handle glucose metabolism may be impaired, leading to an accumulation of glycogen. Earlier studies have shown that there can be a significant increase in glucose storage in the form of glycogen in the kidneys in diabetes. Podocytes play a crucial role in maintaining the integrity of filtration barrier. In diabetes, exposure to elevated glucose levels can lead to significant metabolic and structural changes in podocytes, contributing to kidney damage and the development of diabetic kidney disease. The accumulation of glycogen in podocytes is not a well-established phenomenon. However, a recent study has demonstrated the presence of glycogen granules in podocytes. This review delves into the intricate connections between hyperglycemia and glycogen metabolism within the context of the kidney, with special emphasis on podocytes. The aberrant storage of glycogen has the potential to detrimentally impact podocyte functionality and perturb their structural integrity. This review provides a comprehensive analysis of the alterations in cellular signaling pathways that may potentially lead to glycogen overproduction in podocytes.


Sujet(s)
Néphropathies diabétiques , Hyperglycémie , Podocytes , Humains , Podocytes/métabolisme , Hyperglycémie/métabolisme , Glucose/métabolisme , Néphropathies diabétiques/métabolisme , Glycogène/métabolisme
5.
Biochem Biophys Res Commun ; 679: 145-159, 2023 10 30.
Article de Anglais | MEDLINE | ID: mdl-37696068

RÉSUMÉ

Podocytes are sensitive to insulin, which governs the functional and structural integrity of podocytes that are essential for proper function of the glomerular filtration barrier. Lysosomes are acidic organelles that are implicated in regulation of the insulin signaling pathway. Cathepsin D (CTPD) and lysosome-associated membrane protein 1 (LAMP1) are major lysosomal proteins that reflect the functional state of lysosomes. However, the effect of insulin on lysosome activity and role of lysosomes in the regulation of insulin-dependent glucose uptake in podocytes are unknown. Our studies showed that the short-term incubation of podocytes with insulin decreased LAMP1 and CTPD mRNA levels. Insulin and bafilomycin A1 reduced both the amounts of LAMP1 and CTPD proteins and activity of CTPD, which were associated with a decrease in the fluorescence intensity of lysosomes that were labeled with LysoTracker. Bafilomycin A1 inhibited insulin-dependent endocytosis of the insulin receptor and increased the amounts of the insulin receptor and glucose transporter 4 on the cell surface of podocytes. Bafilomycin A1 also inhibited insulin-dependent glucose uptake despite an increase in the amount of glucose transporter 4 in the plasma membrane of podocytes. These results suggest that lysosomes are signaling hubs that may be involved in the coupling of insulin signaling with the regulation of glucose uptake in podocytes. The dysregulation of this mechanism can lead to the dysfunction of podocytes and development of insulin resistance.


Sujet(s)
Podocytes , Rats , Animaux , Podocytes/métabolisme , Insuline/métabolisme , Récepteur à l'insuline/métabolisme , Facteurs de transcription/métabolisme , Lysosomes/métabolisme , Transduction du signal , Glucose/métabolisme , Transporteurs de glucose par diffusion facilitée/métabolisme
6.
J Cell Physiol ; 238(8): 1921-1936, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37269459

RÉSUMÉ

Podocytes are crucially involved in blood filtration in the glomerulus. Their proper function relies on efficient insulin responsiveness. The insulin resistance of podocytes, defined as a reduction of cell sensitivity to this hormone, is the earliest pathomechanism of microalbuminuria that is observed in metabolic syndrome and diabetic nephropathy. In many tissues, this alteration is mediated by the phosphate homeostasis-controlling enzyme nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1). By binding to the insulin receptor (IR), NPP1 inhibits downstream cellular signaling. Our previous research found that hyperglycemic conditions affect another protein that is involved in phosphate balance, type III sodium-dependent phosphate transporter 1 (Pit 1). In the present study, we evaluated the insulin resistance of podocytes after 24 h of incubation under hyperinsulinemic conditions. Thereafter, insulin signaling was inhibited. The formation of NPP1/IR complexes was observed at that time. A novel finding in the present study was our observation of an interaction between NPP1 and Pit 1 after the 24 h stimulation of podocytes with insulin. After downregulation of the SLC20A1 gene, which encodes Pit 1, we established insulin resistance in podocytes that were cultured under native conditions, manifested as a lack of intracellular insulin signaling and the inhibition of glucose uptake via the glucose transporter type 4. These findings suggest that Pit 1 might be a major factor that participates in the NPP1-mediated inhibition of insulin signaling.


Sujet(s)
Néphropathies diabétiques , Insulinorésistance , Podocytes , Humains , Insuline/pharmacologie , Insuline/métabolisme , Podocytes/métabolisme , Phosphodiesterases/génétique , Phosphodiesterases/métabolisme , Néphropathies diabétiques/métabolisme , Phosphates/métabolisme , Glucose/métabolisme , Cotransporteurs sodium-phosphate de type III/métabolisme
7.
Int J Mol Sci ; 24(4)2023 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-36835364

RÉSUMÉ

The permeability of the glomerular filtration barrier (GFB) is mainly regulated by podocytes and their foot processes. Protein kinase G type Iα (PKGIα) and adenosine monophosphate-dependent kinase (AMPK) affect the contractile apparatus of podocytes and influence the permeability of the GFB. Therefore, we studied the interplay between PKGIα and AMPK in cultured rat podocytes. The glomerular permeability to albumin and transmembrane FITC-albumin flux decreased in the presence of AMPK activators and increased in the presence of PKG activators. The knockdown of PKGIα or AMPK with small-interfering RNA (siRNA) revealed a mutual interaction between PKGIα and AMPK and influenced podocyte permeability to albumin. Moreover, PKGIα siRNA activated the AMPK-dependent signaling pathway. AMPKα2 siRNA increased basal levels of phosphorylated myosin phosphate target subunit 1 and decreased the phosphorylation of myosin light chain 2. Podocytes that were treated with AMPK or PKG activators were characterized by the different organization of actin filaments within the cell. Our findings suggest that mutual interactions between PKGIα and AMPKα2 regulate the contractile apparatus and permeability of the podocyte monolayer to albumin. Understanding this newly identified molecular mechanism in podocytes provides further insights into the pathogenesis of glomerular disease and novel therapeutic targets for glomerulopathies.


Sujet(s)
Albumines , Cyclic GMP-dependent protein kinase type I , Podocytes , Animaux , Rats , AMP/métabolisme , AMP-Activated Protein Kinases/métabolisme , Cyclic GMP-dependent protein kinase type I/métabolisme , Perméabilité , Podocytes/métabolisme , Rat Wistar , Petit ARN interférent/métabolisme , Transduction du signal , Albumines/métabolisme
8.
Cell Signal ; 105: 110622, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36754339

RÉSUMÉ

A decrease in intracellular levels of 3',5'-cyclic guanosine monophosphate (cGMP) has been implicated in the progression of diabetic nephropathy. Hyperglycemia significantly inhibits cGMP-dependent pathway activity in the kidney, leading to glomerular damage and proteinuria. The enhancement of activity of this pathway that is associated with an elevation of cGMP levels may be achieved by inhibition of the cGMP specific phosphodiesterase 5A (PDE5A) using selective inhibitors, such as tadalafil. Hyperglycemia decreased the insulin responsiveness of podocytes and impaired podocyte function. These effects were associated with lower protein amounts and activity of the protein deacetylase sirtuin 1 (SIRT1) and a decrease in the phosphorylation of adenosine monophosphate-dependent protein kinase (AMPK). We found that PDE5A protein levels increased in hyperglycemia, and PDE5A downregulation improved the insulin responsiveness of podocytes with reestablished SIRT1 expression and activity. PDE5A inhibitors potentiate nitric oxide (NO)/cGMP signaling, and NO modulates the activity and expression of SIRT1. Therefore, we investigated the effects of tadalafil on SIRT1 and AMPK in the context of improving the insulin sensitivity in podocytes and podocyte function in hyperglycemia. Our study revealed that tadalafil restored SIRT1 expression and activity and activated AMPK by increasing its phosphorylation. Tadalafil also restored stimulating effect of insulin on glucose transport in podocytes with high glucose-induced insulin resistance. Additionally, tadalafil improved the function of podocytes that were exposed to high glucose concentrations. Our results display novel mechanisms involved in the pathogenesis of glomerulopathies in diabetes, which may contribute to the development of more effective treatment strategies for diabetic nephropathy.


Sujet(s)
Néphropathies diabétiques , Hyperglycémie , Insulinorésistance , Podocytes , Humains , Tadalafil/pharmacologie , Tadalafil/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 5/métabolisme , Insuline/métabolisme , Sirtuine-1/métabolisme , Podocytes/métabolisme , Néphropathies diabétiques/anatomopathologie , AMP-Activated Protein Kinases/métabolisme , GMP cyclique/métabolisme , Glucose/métabolisme
9.
Eur J Cell Biol ; 102(2): 151298, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-36805821

RÉSUMÉ

Lactate has long been acknowledged to be a metabolic waste product, but it has more recently been found as a fuel energy source in mammalian cells. Podocytes are an important component of the glomerular filter, and their role in maintaining the structural integrity of this structure was established. These cells rely on a constant energy supply and reservoir. The utilization of alternative energy substrates to preserve energetic homeostasis is a subject of extensive research, and lactate appears to be one such candidate. Therefore, we investigated the role of lactate as an energy substrate and characterize the lactate transport system in cultured rat podocytes during sufficient and insufficient glucose supplies. The present study, for the first time, demonstrated the presence of lactate transporters in podocytes. Moreover, we observed modified the amount of these transporters in response to limited glucose availability and after l-lactate supplementation. Simultaneously, exposure to l-lactate preserved cell survival during insufficient glucose supply. Interestingly, during glucose deprivation, lactate exposure allowed the steady flow of glycolysis and prevented glycogen reserves depletion. Summarizing, podocytes utilize lactate as an energy substrate and possess a developed system that controls lactate homeostasis, suggesting that it plays an essential role in podocyte metabolism, especially during fluctuations of energy availability.


Sujet(s)
Glucose , Podocytes , Rats , Animaux , Glucose/métabolisme , Podocytes/métabolisme , Glycolyse/physiologie , Acide lactique/métabolisme , Hypoxie cellulaire/physiologie , Mammifères/métabolisme
10.
Sci Rep ; 13(1): 766, 2023 01 14.
Article de Anglais | MEDLINE | ID: mdl-36641502

RÉSUMÉ

Podocytes constitute an external layer of the glomerular filtration barrier, injury to which is a hallmark of renal disease. Mitochondrial dysfunction often accompanies podocyte damage and is associated with an increase in oxidative stress and apoptosis. ß-Aminoisobutyric acid (BAIBA) belongs to natural ß-amino acids and is known to exert anti-inflammatory and antioxidant effects. BAIBA has been reported to be involved in regulating mitochondrial dynamics, but unknown is whether BAIBA influences podocyte bioenergetics. The present study showed that human podocytes express the BAIBA receptor, Mas-related G protein-coupled receptor type D (MRGPRD), which is sensitive to BAIBA stimulation. The treatment of podocytes with L-BAIBA significantly increased their respiratory parameters, such as basal and maximal respiration, adenosine triphosphate (ATP) production, and spare respiratory capacity. We also found that L-BAIBA altered mitochondrial quantity, size, and shape, promoting organelle elongation and branching. L-BAIBA significantly upregulated peroxisome proliferator activated receptor γ coactivator-1α (PGC-1α) and transcription factor A mitochondrial (TFAM), indicating an increase in mitochondrial biogenesis. Our results demonstrate a novel regulatory mechanism of mitochondrial dynamics in podocytes, which may be important for maintaining their functions in the renal filtration barrier and prompting further investigations of preventing or ameliorating mitochondrial damage in podocytes in pathological states.


Sujet(s)
Podocytes , Humains , Podocytes/métabolisme , Biogenèse des organelles , Stress oxydatif , Respiration , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme
11.
Biochim Biophys Acta Mol Cell Res ; 1869(12): 119362, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36152759

RÉSUMÉ

Hyperglycemia significantly decreases 3',5'-cyclic guanosine monophosphate (cGMP)-dependent pathway activity in the kidney. A well-characterized downstream signaling effector of cGMP is cGMP-dependent protein kinase G (PKG), exerting a wide range of downstream effects, including vasodilation and vascular smooth muscle cells relaxation. In podocytes that are exposed to high glucose concentrations, crosstalk between the protein deacetylase sirtuin 1 (SIRT1) and adenosine monophosphate-dependent protein kinase (AMPK) decreased, attenuating insulin responsiveness and impairing podocyte function. The present study examined the effect of enhancing cGMP-dependent pathway activity on SIRT1-AMPK crosstalk in podocytes under hyperglycemic conditions. We found that enhancing cGMP-dependent pathway activity using a cGMP analog was associated with increases in SIRT1 protein levels and activity, with a concomitant increase in the degree of AMPK phosphorylation. The beneficial effects of enhancing cGMP-dependent pathway activity on SIRT1-AMPK crosstalk also included improvements in podocyte function. Based on our findings, we postulate an important role for SIRT1-AMPK crosstalk in the regulation of albumin permeability in hyperglycemia that is strongly associated with activity of the cGMP-dependent pathway.


Sujet(s)
Hyperglycémie , Podocytes , AMP-Activated Protein Kinases/métabolisme , AMP/métabolisme , AMP/pharmacologie , Albumines/métabolisme , Albumines/pharmacologie , Cyclic GMP-Dependent Protein Kinases/métabolisme , Cyclic GMP-Dependent Protein Kinases/pharmacologie , Glucose/métabolisme , Glucose/pharmacologie , Guanosine monophosphate/métabolisme , Guanosine monophosphate/pharmacologie , Humains , Hyperglycémie/métabolisme , Insuline/métabolisme , Phosphorylation , Podocytes/métabolisme , Sirtuine-1/génétique , Sirtuine-1/métabolisme
12.
Biochim Biophys Acta Mol Cell Res ; 1869(9): 119301, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35642843

RÉSUMÉ

Podocyte foot processes are an important cellular layer of the glomerular barrier that regulates glomerular permeability. Insulin via the protein kinase G type Iα (PKGIα) signaling pathway regulates the balance between contractility and relaxation (permeability) of the podocyte barrier by regulation of the actin cytoskeleton. This mechanism was shown to be disrupted in diabetes. Rho family guanosine-5'-triphosphates (GTPases) are dynamic modulators of the actin cytoskeleton and expressed in cells that form the glomerular filtration barrier. Thus, changes in Rho GTPase activity may affect glomerular permeability to albumin. The present study showed that Rho family GTPases control podocyte migration and permeability. Moreover these processes are regulated by insulin in PKGIα-dependent manner. Modulation of the PKGI-dependent activity of Rac1 and RhoA GTPases with inhibitors or small-interfering RNA impair glomerular permeability to albumin. We also demonstrated this mechanism in obese, insulin-resistant Zucker rats. We propose that PKGIα-Rac1-RhoA crosstalk is necessary in proper organization of the podocyte cytoskeleton and consequently the stabilization of glomerular architecture and regulation of filtration barrier permeability.


Sujet(s)
Cyclic GMP-dependent protein kinase type I , Podocytes , Albumines/métabolisme , Animaux , Cyclic GMP-dependent protein kinase type I/métabolisme , Cytosquelette/métabolisme , Insuline/métabolisme , Perméabilité , Podocytes/métabolisme , Rats , Rat Wistar , Rat Zucker , Protéine G rac1/métabolisme , Protéines G rho/métabolisme , Protéine G RhoA/métabolisme
13.
J Mol Med (Berl) ; 100(6): 903-915, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35534645

RÉSUMÉ

Alterations of insulin signaling in diabetes are associated with podocyte injury, proteinuria, and renal failure. Insulin stimulates glucose transport to cells and regulates other intracellular processes that are linked to cellular bioenergetics, such as autophagy, gluconeogenesis, fatty acid metabolism, and mitochondrial homeostasis. The dysfunction of mitochondrial dynamics, including mitochondrial fusion, fission, and mitophagy, has been observed in high glucose-treated podocytes and renal cells from patients with diabetes. Previous studies showed that prolonged hyperglycemia is associated with the development of insulin resistance in podocytes, and high glucose-treated podocytes exhibit an increase in mitochondrial fission and decrease in markers of mitophagy. In the present study, we found that deficiency of the main mitophagy protein PTEN-induced kinase 1 (PINK1) significantly increased albumin permeability and hampered glucose uptake to podocytes. We suggest that PINK1 inhibition impairs the insulin signaling pathway, in which lower levels of phosphorylated Akt and membrane fractions of the insulin receptor and glucose transporter-4 were observed. Moreover, PINK1-depleted podocytes exhibited lower podocin and nephrin expression, thus identifying a potential mechanism whereby albumin leakage increases under hyperglycemic conditions when mitophagy is inhibited. In conclusion, we found that PINK1 plays an essential role in insulin signaling and the maintenance of proper permeability in podocytes. Therefore, PINK1 may be a potential therapeutic target for the treatment or prevention of diabetic nephropathy.


Sujet(s)
Néphropathies diabétiques , Hyperglycémie , Podocytes , Protein kinases , Albumines/métabolisme , Néphropathies diabétiques/métabolisme , Glucose/métabolisme , Humains , Hyperglycémie/métabolisme , Insuline/métabolisme , Phosphohydrolase PTEN/métabolisme , Perméabilité , Podocytes/métabolisme , Protein kinases/génétique , Protein kinases/métabolisme , Transduction du signal
14.
J Cell Physiol ; 237(5): 2478-2491, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35150131

RÉSUMÉ

Soft tissue calcification is a pathological phenomenon that often occurs in end-stage chronic kidney disease (CKD), which is caused by diabetic nephropathy, among other factors. Hyperphosphatemia present during course of CKD contributes to impairments in kidney function, particularly damages in the glomerular filtration barrier (GFB). Essential elements of the GFB include glomerular epithelial cells, called podocytes. In the present study, we found that human immortalized podocytes express messenger RNA and protein of phosphate transporters, including NaPi 2c (SLC34A3), Pit 1 (SLC20A1), and Pit 2 (SLC20A2), which are sodium-dependent and mediate intracellular phosphate (Pi) transport, and XPR1, which is responsible for extracellular Pi transport. We found that cells that were grown in a medium with a high glucose (HG) concentration (30 mM) expressed less Pit 1 and Pit 2 protein than podocytes that were cultured in a standard glucose medium (11 mM). We found that exposure of the analyzed transporters in the cell membrane of the podocyte is altered by HG conditions. We also found that the activity of tissue nonspecific alkaline phosphatase increased in HG, causing a rise in Pi generation. Additionally, HG led to a reduction of the amount of ectonucleotide pyrophosphatase/phosphodiesterase 1 in the cell membrane of podocytes. The extracellular concentration of pyrophosphate also decreased under HG conditions. These data suggest that a hyperglycemic environment enhances the production of Pi in podocytes and its retention in the extracellular space, which may induce glomerular calcification.


Sujet(s)
Calcinose , Podocytes , Insuffisance rénale chronique , Calcinose/métabolisme , Glucose/métabolisme , Humains , Glomérule rénal/métabolisme , Protéines de transport du phosphate/génétique , Protéines de transport du phosphate/métabolisme , Podocytes/métabolisme , Insuffisance rénale chronique/anatomopathologie , Cotransporteurs sodium-phosphate de type III/génétique , Cotransporteurs sodium-phosphate de type III/métabolisme
15.
Arch Biochem Biophys ; 714: 109078, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34742673

RÉSUMÉ

The purinergic activation of P2 receptors initiates a powerful and rapid signaling cascade that contributes to the regulation of an array of physiological and pathophysiological processes in many organs, including the kidney. P2 receptors are broadly distributed in both epithelial and vascular renal cells. Disturbances of purinergic signaling can lead to impairments in renal function. A growing body of evidence indicates changes in P2 receptor expression and nucleotide metabolism in chronic renal injury and inflammatory diseases. Increasing attention has focused on purinergic P2X7 receptors, which are not normally expressed in healthy kidney tissue but are highly expressed at sites of tissue damage and inflammation. Under hyperglycemic conditions, several mechanisms that are linked to purinergic signaling and involve nucleotide release and degradation are disrupted, resulting in the accumulation of adenosine 5'-triphosphate in the bloodstream in diabetes. Dysfunction of the purinergic system might be associated with serious vascular complications in diabetes, including diabetic nephropathy. This review summarizes our current knowledge of the role of P2 receptors in diabetes-related glomerular injury and its implications for new therapeutics for diabetic nephropathy.


Sujet(s)
Néphropathies diabétiques/métabolisme , Glomérule rénal/métabolisme , Glomérule rénal/anatomopathologie , Récepteurs purinergiques P2/métabolisme , Animaux , Humains , Transduction du signal
16.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-34360633

RÉSUMÉ

Hyperglycemic conditions (HG), at early stages of diabetic nephropathy (DN), cause a decrease in podocyte numbers and an aberration of their function as key cells for glomerular plasma filtration. Klotho protein was shown to overcome some negative effects of hyperglycemia. Klotho is also a coreceptor for fibroblast growth factor receptors (FGFRs), the signaling of which, together with a proper rate of glycolysis in podocytes, is needed for a proper function of the glomerular filtration barrier. Therefore, we measured levels of Klotho in renal tissue, serum, and urine shortly after DN induction. We investigated whether it influences levels of FGFRs, rates of glycolysis in podocytes, and albumin permeability. During hyperglycemia, the level of membrane-bound Klotho in renal tissue decreased, with an increase in the shedding of soluble Klotho, its higher presence in serum, and lower urinary excretion. The addition of Klotho increased FGFR levels, especially FGFR1/FGFR2, after their HG-induced decrease. Klotho also increased levels of glycolytic parameters of podocytes, and decreased podocytic and glomerular albumin permeability in HG. Thus, we found that the decrease in the urinary excretion of Klotho might be an early biomarker of DN and that Klotho administration may have several beneficial effects on renal function in DN.


Sujet(s)
Glucuronidase/métabolisme , Hyperglycémie/métabolisme , Podocytes/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Animaux , Glycolyse , Protéines Klotho , Mâle , Perméabilité , Rat Wistar
17.
Arch Biochem Biophys ; 710: 109005, 2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34371008

RÉSUMÉ

Glomerular podocytes are a target for the actions of insulin. Accumulating evidence indicates that exposure to nutrient overload induces insulin resistance in these cells, manifested by abolition of the stimulatory effect of insulin on glucose uptake. Numerous recent studies have investigated potential mechanisms of the induction of insulin resistance in podocytes. High glucose concentrations stimulated reactive oxygen species production through NADPH oxidase activation, decreased adenosine monophosphate-activated protein kinase (AMPK) phosphorylation, and reduced deacetylase sirtuin 1 (SIRT1) protein levels and activity. Calcium signaling involving transient receptor potential cation channel C, member 6 (TRPC6) also was demonstrated to play an essential role in the regulation of insulin-dependent signaling and glucose uptake in podocytes. Furthermore, podocytes exposed to diabetic environment, with elevated insulin levels become insulin resistant as a result of degradation of insulin receptor (IR), resulting in attenuation of insulin signaling responsiveness. Also elevated levels of palmitic acid appear to be an important factor and contributor to podocytes insulin resistance. This review summarizes cellular and molecular alterations that contribute to the development of insulin resistance in glomerular podocytes.


Sujet(s)
Insulinorésistance/physiologie , Podocytes/métabolisme , AMP-Activated Protein Kinases/métabolisme , Animaux , Transport biologique actif , Signalisation calcique , Néphropathies diabétiques/métabolisme , Stress du réticulum endoplasmique , Glucose/métabolisme , Hexosamine/biosynthèse , Humains , Voies et réseaux métaboliques , Modèles biologiques , Phosphorylation , Espèces réactives de l'oxygène/métabolisme , Récepteur à l'insuline/métabolisme , Transduction du signal , Sirtuine-1/métabolisme , Réponse aux protéines mal repliées
18.
Exp Cell Res ; 407(1): 112758, 2021 10 01.
Article de Anglais | MEDLINE | ID: mdl-34437881

RÉSUMÉ

Podocytes constitute the outer layer of the renal glomerular filtration barrier. Their energy requirements strongly depend on efficient oxidative respiration, which is tightly connected with mitochondrial dynamics. We hypothesized that hyperglycemia modulates energy metabolism in glomeruli and podocytes and contributes to the development of diabetic kidney disease. We found that oxygen consumption rates were severely reduced in glomeruli from diabetic rats and in human podocytes that were cultured in high glucose concentration (30 mM; HG). In these models, all of the mitochondrial respiratory parameters, including basal and maximal respiration, ATP production, and spare respiratory capacity, were significantly decreased. Podocytes that were treated with HG showed a fragmented mitochondrial network, together with a decrease in expression of the mitochondrial fusion markers MFN1, MFN2, and OPA1, and an increase in the activity of the fission marker DRP1. We showed that markers of mitochondrial biogenesis, such as PGC-1α and TFAM, decreased in HG-treated podocytes. Moreover, PINK1/parkin-dependent mitophagy was inhibited in these cells. These results provide evidence that hyperglycemia impairs mitochondrial dynamics and turnover, which may underlie the remarkable deterioration of mitochondrial respiration parameters in glomeruli and podocytes.


Sujet(s)
Hyperglycémie/métabolisme , Mitochondries/métabolisme , Mitophagie/physiologie , Podocytes/métabolisme , Animaux , Diabète expérimental/métabolisme , Humains , Rein/métabolisme , Mâle , Protein kinases/métabolisme , Rat Wistar , Espèces réactives de l'oxygène/métabolisme
19.
Biochem Pharmacol ; 192: 114687, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34274355

RÉSUMÉ

Podocytes and their foot processes form an important cellular layer of the glomerular barrier involved in regulating glomerular permeability. Disturbances in podocyte function play a central role in the development of proteinuria in diabetic nephropathy. The retraction of podocyte foot processes forming a slit diaphragm is a common feature of proteinuria. Metformin is an oral antidiabetic agent of the biguanide class that is widely recommended for the treatment of high blood glucose in patients with type 2 diabetes mellitus. In addition to lowering glucose, several recent studies have reported potential beneficial effects of metformin on diabetic kidney function. Furthermore, a key molecule of the antidiabetic mechanism of action of metformin is adenosine 5'-monophospate-activated protein kinase (AMPK), as the metformin-induced activation of AMPK is well documented. The present review summarizes current knowledge on the protective effects of metformin against pathological changes in podocytes that are induced by hyperglycemia.


Sujet(s)
Diabète/traitement médicamenteux , Diabète/métabolisme , Hypoglycémiants/usage thérapeutique , Metformine/usage thérapeutique , Podocytes/métabolisme , AMP-Activated Protein Kinases/métabolisme , Animaux , Humains , Hypoglycémiants/pharmacologie , Insuline/métabolisme , Metformine/pharmacologie , Podocytes/effets des médicaments et des substances chimiques , Résultat thérapeutique
20.
Arch Biochem Biophys ; 709: 108985, 2021 09 30.
Article de Anglais | MEDLINE | ID: mdl-34252390

RÉSUMÉ

The protein deacetylase sirtuin 1 (SIRT1) and adenosine monophosphate-dependent protein kinase (AMPK) play important roles in the development of insulin resistance. In glomerular podocytes, crosstalk between these two enzymes may be altered under hyperglycemic conditions. SIRT1 protein levels and activity and AMPK phosphorylation decrease under hyperglycemic conditions, with concomitant inhibition of the effect of insulin on glucose uptake into these cells. Nitric oxide (NO)-dependent regulatory signaling pathways have been shown to be downregulated under diabetic conditions. The present study examined the involvement of the NO synthase (NOS)/NO pathway in the regulation of SIRT1-AMPK signaling and glucose uptake in podocytes. We examined the effects of NOS/NO pathway alterations on SIRT1/AMPK signaling and glucose uptake using pharmacological tools and a small-interfering transfection approach. We also examined the ability of the NOS/NO pathway to protect podocytes against high glucose-induced alterations of SIRT1/AMPK signaling and insulin-dependent glucose uptake. Inhibition of the NOS/NO pathway reduced SIRT1 protein levels and activity, leading to a decrease in AMPK phosphorylation and blockade of the effect of insulin on glucose uptake. Treatment with the NO donor S-nitroso-N-acetylpenicillamine (SNAP) prevented high glucose-induced decreases in SIRT1 and AMPK activity and increased GLUT4 protein expression, thereby improving glucose uptake in podocytes. These findings suggest that inhibition of the NOS/NO pathway may result in alterations of the effects of insulin on glucose uptake in podocytes. In turn, the enhancement of NOS/NO pathway activity may prevent these deleterious effects of high glucose concentrations, thus bidirectionally stimulating the SIRT1-AMPK reciprocal activation loop.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Glucose/métabolisme , Nitric oxide synthase type I/métabolisme , Monoxyde d'azote/métabolisme , Podocytes/métabolisme , Sirtuine-1/métabolisme , Animaux , Régulation négative/physiologie , Antienzymes/pharmacologie , Techniques de knock-down de gènes , Transporteur de glucose de type 4/métabolisme , Insuline/métabolisme , Insulinorésistance/physiologie , L-NAME/pharmacologie , Nitric oxide synthase type I/antagonistes et inhibiteurs , Phosphorylation/effets des médicaments et des substances chimiques , Rats , N-Acétyl-S-nitroso-pénicillamine/pharmacologie , Transduction du signal , Sirtuine-1/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...