Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 105(1): 54-64, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38707675

RESUMEN

The neonatal Fc receptor (FcRn) was initially discovered as the receptor that allowed passive immunity in newborns by transporting maternal IgG through the placenta and enterocytes. Since its initial discovery, FcRn has been found to exist throughout all stages of life and in many different cell types. Beyond passive immunity, FcRn is necessary for intrinsic albumin and IgG recycling and is important for antigen processing and presentation. Given its multiple important roles, FcRn has been utilized in many disease treatments including a new class of agents that were developed to inhibit FcRn for treatment of a variety of autoimmune diseases. Certain cell populations within the kidney also express high levels of this receptor. Specifically, podocytes, proximal tubule epithelial cells, and vascular endothelial cells have been found to utilize FcRn. In this review, we summarize what is known about FcRn and its function within the kidney. We also discuss how FcRn has been used for therapeutic benefit, including how newer FcRn inhibiting agents are being used to treat autoimmune diseases. Lastly, we will discuss what renal diseases may respond to FcRn inhibitors and how further work studying FcRn within the kidney may lead to therapies for kidney diseases.


Asunto(s)
Antígenos de Histocompatibilidad Clase I , Enfermedades Renales , Receptores Fc , Humanos , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Receptores Fc/metabolismo , Receptores Fc/inmunología , Receptores Fc/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/terapia , Enfermedades Renales/inmunología , Animales , Riñón/metabolismo , Riñón/inmunología , Riñón/patología , Podocitos/metabolismo , Podocitos/inmunología , Inmunoglobulina G/metabolismo , Inmunoglobulina G/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo
2.
J Am Soc Nephrol ; 33(8): 1477-1486, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35459732

RESUMEN

BACKGROUND: The proximal tubules play a critical role in phosphate (Pi) homeostasis by reabsorbing Pi via sodium-dependent Pi cotransporters. NPT2A is a major proximal-specific Pi cotransporter, whose expression is regulated by circulating hormones, such as parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23). In this study, we aimed to find a novel regulator in Pi homeostasis. METHODS: Using RNA-seq and RT-qPCR analysis, we identified proximal tubule cell-enriched genes. We next used RNAi screening of the identified proximal tubular cell-enriched genes to identify a novel proximal tubule-specific gene that contributes to FGF23- and PTH-mediated inhibition of Pi uptake and NPT2 reduction. We created mice lacking this novel regulator of Pi homeostasis to examine whether the novel regulator contributes to Pi homeostasis in vivo. RESULTS: We identified 54 kidney-enriched genes, 19 of which are expressed in renal primary proximal tubule cells. One of the proximal tubule-specific genes, TMEM174, interacted with NPT2A, and its knockdown blocked the reduction of NPT2A protein by FGF23 and PTH treatments in human and opossum proximal tubule cells. TMEM174 KO mice had significantly increased levels of serum Pi, FGF23, and PTH, resulting in vascular calcification. CONCLUSIONS: TMEM174 is a novel regulator of Pi homeostasis that interacts with NPT2A.


Asunto(s)
Hiperfosfatemia , Proteínas de la Membrana , Calcificación Vascular , Animales , Factores de Crecimiento de Fibroblastos , Humanos , Hiperfosfatemia/genética , Túbulos Renales Proximales/metabolismo , Proteínas de la Membrana/genética , Ratones , Hormona Paratiroidea , Fosfatos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Calcificación Vascular/genética
3.
Kidney Int ; 102(6): 1331-1344, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36063874

RESUMEN

Complement activation at a particular location is determined by the balance of activating and inhibitory proteins. Factor H is a key regulator of the alternative pathway of complement, and genetic or acquired impairments in Factor H are associated with glomerular injury. The human Factor H-related proteins (FHRs) comprise a family of five proteins that are structurally related to Factor H. Variations in the genes or expression levels of the FHRs are also associated with glomerular disease, although the mechanisms of glomerular protection/injury are incompletely understood. To explore the role of the FHRs on complement regulation/dysregulation in the kidney, we expressed and purified recombinant murine FHRs (FHRs A, B, C and E). These four distinct FHRs contain binding regions with high amino acid sequence homology to binding regions within Factor H, but we observed different interactions of the FHRs with Factor H binding ligands, including heparin and C3d. There was differential binding of the FHRs to the resident kidney cell types (mesangial, glomerular endothelial, podocytes, and tubular epithelial). All four FHRs caused complement dysregulation on kidney cell surfaces in vitro, although the magnitude of the effect differed among the FHRs and also varied among the different kidney cells. However, only FHR E caused glomerular complement dysregulation when injected in vivo but did not exacerbate injury when injected into mice with ischemic acute kidney injury, an alternative pathway-mediated model. Thus, our experiments demonstrate that the FHRs have unique, and likely context-dependent, effects on the different cell types within the kidney.


Asunto(s)
Factor H de Complemento , Enfermedades Renales , Humanos , Ratones , Animales , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Riñón/metabolismo
4.
Kidney Int ; 97(5): 966-979, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32081304

RESUMEN

Neutrophil gelatinase associated lipocalin (NGAL, Lcn2) is the most widely studied biomarker of acute kidney injury (AKI). Previous studies have demonstrated that NGAL is produced by the kidney and released into the urine and plasma. Consequently, NGAL is currently considered a tubule specific injury marker of AKI. However, the utility of NGAL to predict AKI has been variable suggesting that other mechanisms of production are present. IL-6 is a proinflammatory cytokine increased in plasma by two hours of AKI and mediates distant organ effects. Herein, we investigated the role of IL-6 in renal and extra-renal NGAL production. Wild type mice with ischemic AKI had increased plasma IL-6, increased hepatic NGAL mRNA, increased plasma NGAL, and increased urine NGAL; all reduced in IL-6 knockout mice. Intravenous IL-6 in normal mice increased hepatic NGAL mRNA, plasma NGAL and urine NGAL. In mice with hepatocyte specific NGAL deletion (Lcn2hep-/-) and ischemic AKI, hepatic NGAL mRNA was absent, and plasma and urine NGAL were reduced. Since urine NGAL levels appear to be dependent on plasma levels, the renal handling of circulating NGAL was examined using recombinant human NGAL. After intravenous recombinant human NGAL administration to mice, human NGAL in mouse urine was detected by ELISA during proximal tubular dysfunction, but not in pre-renal azotemia. Thus, during AKI, IL-6 mediates hepatic NGAL production, hepatocytes are the primary source of plasma and urine NGAL, and plasma NGAL appears in the urine during proximal tubule dysfunction. Hence, our data change the paradigm by which NGAL should be interpreted as a biomarker of AKI.


Asunto(s)
Lesión Renal Aguda , Lipocalinas , Lesión Renal Aguda/diagnóstico , Proteínas de Fase Aguda/genética , Animales , Biomarcadores , Hepatocitos , Interleucina-6 , Lipocalina 2/genética , Ratones
5.
Am J Physiol Cell Physiol ; 317(5): C1048-C1060, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31553647

RESUMEN

The neonatal Fc receptor (FcRn) has been shown to be required for antigen presentation in dendritic cells, and global knockout of FcRn attenuates immune-mediated kidney disease. Podocytes express interleukin-6 (IL-6) receptor and produce IL-6 under proinflammatory conditions. Here we examined the role of FcRn in the IL-6-mediated inflammatory response in podocytes. We examined IL-6 production by ELISA and expression by qPCR in wild type (WT) and FcRn knockout (KO) podocytes after treatment with proinflammatory stimuli as well as IL-6-mediated signaling via the JAK/STAT pathway. We also examined podocyte motility in cultured WT and KO podocytes after a proinflammatory challenge. We found that FcRn KO podocytes produced minimal amount of IL-6 after treatment with albumin, IgG, or immune complexes whereas WT podocytes had a robust response. FcRn KO podocytes also had minimal expression of IL-6 compared with WT. By Western blotting, there was significantly less phosphorylated STAT3 in KO podocytes after treatment with IFNγ or immune complexes. In a scratch assay, FcRn KO podocytes showed increased motility comparted KO, suggesting a defect in actin dynamics. Cultured FcRn KO podocytes also demonstrated abnormal stress fibers compared with WT and the defect could be rescued by IL-6 treatment. This study shows that in podocytes, FcRn modulates the IL-6 mediated response to proinflammatory stimuli and regulates podocytes actin structure, motility and synaptopodin expression.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Interleucina-6/metabolismo , Podocitos/metabolismo , Receptores Fc/deficiencia , Transducción de Señal/fisiología , Citoesqueleto de Actina/genética , Animales , Línea Celular Transformada , Células Cultivadas , Antígenos de Histocompatibilidad Clase I/genética , Interleucina-6/genética , Ratones , Ratones Noqueados , Receptores Fc/genética
6.
Am J Physiol Renal Physiol ; 306(9): F941-51, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24573386

RESUMEN

The renal glomerulus forms a selective filtration barrier that allows the passage of water, ions, and small solutes into the urinary space while restricting the passage of cells and macromolecules. The three layers of the glomerular filtration barrier include the vascular endothelium, glomerular basement membrane (GBM), and podocyte epithelium. Podocytes are capable of internalizing albumin and are hypothesized to clear proteins that traverse the GBM. The present study followed the fate of FITC-labeled albumin to establish the mechanisms of albumin endocytosis and processing by podocytes. Confocal imaging and total internal reflection fluorescence microscopy of immortalized human podocytes showed FITC-albumin endocytosis occurred preferentially across the basal membrane. Inhibition of clathrin-mediated endocytosis and caveolae-mediated endocytosis demonstrated that the majority of FITC-albumin entered podocytes through caveolae. Once internalized, FITC-albumin colocalized with EEA1 and LAMP1, endocytic markers, and with the neonatal Fc receptor, a marker for transcytosis. After preloading podocytes with FITC-albumin, the majority of loaded FITC-albumin was lost over the subsequent 60 min of incubation. A portion of the loss of albumin occurred via lysosomal degradation as pretreatment with leupeptin, a lysosomal protease inhibitor, partially inhibited the loss of FITC-albumin. Consistent with transcytosis of albumin, preloaded podocytes also progressively released FITC-albumin into the extracellular media. These studies confirm the ability of podocytes to endocytose albumin and provide mechanistic insight into cellular mechanisms and fates of albumin handling in podocytes.


Asunto(s)
Caveolas/metabolismo , Endocitosis , Fluoresceína-5-Isotiocianato/análogos & derivados , Podocitos/metabolismo , Albúmina Sérica/metabolismo , Animales , Biomarcadores/metabolismo , Caveolas/efectos de los fármacos , Línea Celular , Polaridad Celular , Endocitosis/efectos de los fármacos , Fluoresceína-5-Isotiocianato/metabolismo , Tasa de Filtración Glomerular , Humanos , Cinética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/enzimología , Masculino , Podocitos/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Transporte de Proteínas , Proteolisis , Ratas , Ratas Sprague-Dawley , Receptores Fc/metabolismo , Transcitosis , Proteínas de Transporte Vesicular/metabolismo
7.
Cells ; 13(7)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38607033

RESUMEN

Research into the neonatal Fc receptor (FcRn) has increased dramatically ever since Simister and Mostov first purified a rat version of the receptor. Over the years, FcRn has been shown to function not only as a receptor that transfers immunity from mother to fetus but also performs an array of different functions that include transport and recycling of immunoglobulins and albumin in the adult. Due to its important cellular roles, several clinical trials have been designed to either inhibit/enhance FcRn function or develop of non-invasive therapeutic delivery system such as fusion of drugs to IgG Fc or albumin to enhance delivery inside the cells. Here, we report the accidental identification of several FcRn alternatively spliced variants in both mouse and human cells. The four new mouse splice variants are capable of binding immunoglobulins' Fc and Fab portions. In addition, we have identified FcRn-specific vesicles in which immunoglobulins and albumin can be stored and that are involved in the endosomal-lysosomal system. The complexity of FcRn functions offers significant potential to design and develop novel and targeted therapeutics.


Asunto(s)
Receptores Fc , Animales , Humanos , Ratones , Ratas , Albúminas/metabolismo , Endosomas/metabolismo , Inmunoglobulina G/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Isoformas de Proteínas
8.
PLoS One ; 18(4): e0284636, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37071647

RESUMEN

Podocytes are key to preventing the filtration of serum proteins into the urine. Recent evidence also suggests that in immune mediated kidney diseases, podocytes are the targets of immune complexes (ICs). The mechanisms whereby podocytes handle and respond to ICs remain unknown. The neonatal Fc receptor (FcRn) is involved in IgG handling in podocytes and is also required in dendritic cells to traffic ICs to the lysosome for proteolytic degradation of antigen and presentation on MHC II. Here we examine the role of FcRn in handling ICs in podocytes. We show that knockout of FcRn in podocytes results in decreased trafficking of ICs to the lysosome and increases IC trafficking to recycling endosomes. FcRn KO also alters lysosomal distribution, decreases lysosomal surface area and decreases cathepsin B expression and activity. We demonstrate that signaling pathways in cultured podocytes differ after treatment with IgG alone versus ICs and that podocyte proliferation in both WT and KO podocytes is suppressed by IC treatment. Our findings suggest that podocytes respond differentially to IgG versus ICs and that FcRn modifies the lysosomal response to ICs. Elucidating the mechanisms underlying podocyte handling of ICs may provide novel pathways to modulate immune mediated kidney disease progression.


Asunto(s)
Podocitos , Ratones , Animales , Podocitos/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Ratones Noqueados , Inmunoglobulina G , Antígenos de Histocompatibilidad Clase I , Receptores Fc , Lisosomas/metabolismo
9.
J Biol Chem ; 286(17): 15032-42, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21388960

RESUMEN

The sodium-dependent phosphate (Na/P(i)) transporters NaPi-2a and NaPi-2c play a major role in the renal reabsorption of P(i). The functional need for several transporters accomplishing the same role is still not clear. However, the fact that these transporters show differential regulation under dietary and hormonal stimuli suggests different roles in P(i) reabsorption. The pathways controlling this differential regulation are still unknown, but one of the candidates involved is the NHERF family of scaffolding PDZ proteins. We propose that differences in the molecular interaction with PDZ proteins are related with the differential adaptation of Na/P(i) transporters. Pdzk1(-/-) mice adapted to chronic low P(i) diets showed an increased expression of NaPi-2a protein in the apical membrane of proximal tubules but impaired up-regulation of NaPi-2c. These results suggest an important role for PDZK1 in the stabilization of NaPi-2c in the apical membrane. We studied the specific protein-protein interactions of Na/P(i) transporters with NHERF-1 and PDZK1 by FRET. FRET measurements showed a much stronger interaction of NHERF-1 with NaPi-2a than with NaPi-2c. However, both Na/P(i) transporters showed similar FRET efficiencies with PDZK1. Interestingly, in cells adapted to low P(i) concentrations, there were increases in NaPi-2c/PDZK1 and NaPi-2a/NHERF-1 interactions. The differential affinity of the Na/P(i) transporters for NHERF-1 and PDZK1 proteins could partially explain their differential regulation and/or stability in the apical membrane. In this regard, direct interaction between NaPi-2c and PDZK1 seems to play an important role in the physiological regulation of NaPi-2c.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/metabolismo , Adaptación Fisiológica , Animales , Polaridad Celular , Transferencia Resonante de Energía de Fluorescencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón , Proteínas de la Membrana , Ratones , Ratones Noqueados , Unión Proteica , Estabilidad Proteica
10.
Int J Nephrol Renovasc Dis ; 15: 17-25, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35177925

RESUMEN

BACKGROUND: Based on current ISPD guidelines, it is unclear as to whether ceftazidime should be discontinued in subsequent management of culture-negative peritonitis if it is used as empirical gram-negative coverage. Herein, we aim to compare the clinical outcomes of cefazolin plus ceftazidime versus cefazolin alone. METHODS: This was a retrospective cohort study. Adult peritoneal dialysis (PD) patients who were diagnosed with culture-negative peritonitis between 2014 and 2020 were included. Patients were categorized into two groups according to treatment regimen. Primary response rate, peritonitis relapse rate, and time to primary response were compared. Factors that predicted primary response were determined using Cox regression analysis. RESULTS: A total of 58 patients were included in the study. Of these, 42 received cefazolin plus ceftazidime and 16 received cefazolin monotherapy. Overall, the mean age was 65.7±10.4 years. Most of the patients (81.3%) were prescribed continuous ambulatory peritoneal dialysis. Initial effluent WBC was 4211±10357 in the combination group and 3833±6931 cell/mm3 in the monotherapy group (p=0.89). There was no significant difference in primary response at day 5 between the two groups (95.2% in the combination group vs93.7% in the monotherapy group, p=0.82). However, cumulative probability of primary response by the Kaplan-Meier analysis in the combination group was higher than in the monotherapy group (p=0.02). Adjusted HR of serum potassium level to predict a primary response was 1.83 according to multivariate analysis (p=0.03). There was no difference between the two groups in terms of peritonitis relapse or catheter removal. CONCLUSION: This is the first study to compare clinical outcomes between cefazolin plus ceftazidime versus cefazolin monotherapy in culture-negative peritonitis. Our results suggest that if peritonitis is resolving at day 3, discontinuation of ceftazidime could yield favorable treatment outcomes and might be appropriate for subsequent management. However, the risk of not having gram-negative coverage should be considered.

11.
Kidney Int Rep ; 7(4): 797-809, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35497798

RESUMEN

Introduction: Minimal change disease (MCD) is considered a podocyte disorder triggered by unknown circulating factors. Here, we hypothesized that the endothelial cell (EC) is also involved in MCD. Methods: We studied 45 children with idiopathic nephrotic syndrome (44 had steroid sensitive nephrotic syndrome [SSNS], and 12 had biopsy-proven MCD), 21 adults with MCD, and 38 healthy controls (30 children, 8 adults). In circulation, we measured products of endothelial glycocalyx (EG) degradation (syndecan-1, heparan sulfate [HS] fragments), HS proteoglycan cleaving enzymes (matrix metalloprotease-2 [MMP-2], heparanase activity), and markers of endothelial activation (von Willebrand factor [vWF], thrombomodulin) by enzyme-linked immunosorbent assay (ELISA) and mass spectrometry. In human kidney tissue, we assessed glomerular EC (GEnC) activation by immunofluorescence of caveolin-1 (n = 11 MCD, n = 5 controls). In vitro, we cultured immortalized human GEnC with sera from control subjects and patients with MCD/SSNS sera in relapse (n = 5 per group) and performed Western blotting of thrombomodulin of cell lysates as surrogate marker of endothelial activation. Results: In circulation, median concentrations of all endothelial markers were higher in patients with active disease compared with controls and remained high in some patients during remission. In the MCD glomerulus, caveolin-1 expression was higher, in an endothelial-specific pattern, compared with controls. In cultured human GEnC, sera from children with MCD/SSNS in relapse increased thrombomodulin expression compared with control sera. Conclusion: Our data show that alterations involving the systemic and glomerular endothelium are nearly universal in patients with MCD and SSNS, and that GEnC can be directly activated by circulating factors present in the MCD/SSNS sera during relapse.

12.
Am J Physiol Cell Physiol ; 301(4): C850-61, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21593452

RESUMEN

The kidney is a key regulator of phosphate homeostasis. There are two predominant renal sodium phosphate cotransporters, NaPi2a and NaPi2c. Both are regulated by parathyroid hormone (PTH), which decreases the abundance of the NaPi cotransporters in the apical membrane of renal proximal tubule cells. The time course of PTH-induced removal of the two cotransporters from the apical membrane, however, is markedly different for NaPi2a compared with NaPi2c. In animals and in cell culture, PTH treatment results in almost complete removal of NaPi2a from the brush border (BB) within 1 h whereas for NaPi2c this process in not complete until 4 to 8 h after PTH treatment. The reason for this is poorly understood. We have previously shown that the unconventional myosin motor myosin VI is required for PTH-induced removal of NaPi2a from the proximal tubule BB. Here we demonstrate that myosin VI is also necessary for PTH-induced removal of NaPi2c from the apical membrane. In addition, we show that, while at baseline the two cotransporters have similar diffusion coefficients within the membrane, after PTH addition the diffusion coefficient for NaPi2a initially exceeds that for NaPi2c. Thus NaPi2c appears to remain "tethered" in the apical membrane for longer periods of time after PTH treatment, accounting, at least in part, for the difference in response times to PTH of NaPi2a versus NaPi2c.


Asunto(s)
Hormona Paratiroidea/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/metabolismo , Actinas/fisiología , Animales , Línea Celular , Citoesqueleto/fisiología , Túbulos Renales Proximales/citología , Ratones , Ratones Endogámicos C57BL , Microvellosidades/efectos de los fármacos , Microvellosidades/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Zarigüeyas , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/genética
13.
Kidney Int ; 80(5): 535-44, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21677638

RESUMEN

Cholesterol is pumped out of the cells in different tissues, including the vasculature, intestine, liver, and kidney, by the ATP-binding cassette transporters. Ligands that activate the liver X receptor (LXR) modulate this efflux. Here we determined the effects of LXR agonists on the regulation of phosphate transporters. Phosphate homeostasis is regulated by the coordinated action of the intestinal and renal sodium-phosphate (NaPi) transporters, and the loss of this regulation causes hyperphosphatemia. Mice treated with DMHCA or TO901317, two LXR agonists that prevent atherosclerosis in ApoE or LDLR knockout mice, significantly decreased the activity of intestinal and kidney proximal tubular brush border membrane sodium gradient-dependent phosphate uptake, decreased serum phosphate, and increased urine phosphate excretion. The effects of DMHCA were due to a significant decrease in the abundance of the intestinal and renal NaPi transport proteins. The same effect was also found in opossum kidney cells in culture after treatment with either agonist. There was increased nuclear expression of the endogenous LXR receptor, a reduction in NaPi4 protein abundance (the main type II NaPi transporter in the opossum cells), and a reduction in NaPi co-transport activity. Thus, LXR agonists modulate intestinal and renal NaPi transporters and, in turn, serum phosphate levels.


Asunto(s)
Receptores Nucleares Huérfanos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/metabolismo , Animales , Transporte Biológico , Homeostasis , Humanos , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Ligandos , Receptores X del Hígado , Ratones , Receptores Nucleares Huérfanos/agonistas , Fosfatos/metabolismo , Fosfatos/orina
14.
Methods Mol Biol ; 2224: 123-132, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606211

RESUMEN

Proteinuria is a widely used marker of renal disease and is strongly associated with renal and cardiovascular outcomes. The molecular mechanisms underlying filtration of serum proteins through the glomerular filtration barrier (GFB) remain to be determined. Since the GFB is a complex structure, studies of albumin or IgG trafficking in cultured cells in vitro may not fully recapitulate these processes in vivo. In other epithelial cells including renal proximal tubular cells, the neonatal Fc receptor (FcRn) is required to divert albumin and IgG from the degradative pathway which allows these proteins to be recycled or transcytosed. To examine the role of podocyte FcRn in albumin and IgG trafficking in vivo, we detail the creation of a podocyte-specific FcRn knockout mouse and describe methods for examining intraglomerular detection of albumin and IgG in these mice.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Podocitos/metabolismo , Receptores Fc/metabolismo , Albúminas/metabolismo , Animales , Células Epiteliales/metabolismo , Femenino , Inmunoglobulina G/metabolismo , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas/fisiología , Proteinuria/metabolismo , Transcitosis/fisiología
15.
Cell Death Dis ; 12(3): 248, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674554

RESUMEN

Many surgical models are used to study kidney and other diseases in mice, yet the effects of the surgical procedure itself on the kidney and other tissues have not been elucidated. In the present study, we found that both sham surgery and unilateral nephrectomy (UNX), which is used as a model of renal compensatory hypertrophy, in mice resulted in increased mammalian target of rapamycin complex 1/2 (mTORC1/2) in the remaining kidney. mTORC1 is known to regulate lysosomal biogenesis and autophagy. Genes associated with lysosomal biogenesis and function were decreased in sham surgery and UNX kidneys. In both sham surgery and UNX, there was suppressed autophagic flux in the kidney as indicated by the lack of an increase in LC3-II or autophagosomes seen on immunoblot, IF and EM after bafilomycin A1 administration and a concomitant increase in p62, a marker of autophagic cargo. There was a massive increase in pro-inflammatory cytokines, which are known to activate ERK1/2, in the serum after sham surgery and UNX. There was a large increase in ERK1/2 in sham surgery and UNX kidneys, which was blocked by the MEK1/2 inhibitor, trametinib. Trametinib also resulted in a significant decrease in p62. In summary, there was an intense systemic inflammatory response, an ERK-mediated increase in p62 and suppressed autophagic flux in the kidney after sham surgery and UNX. It is important that researchers are aware that changes in systemic pro-inflammatory cytokines, ERK1/2 and autophagy can be caused by sham surgery as well as the kidney injury/disease itself.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Enfermedades Renales/metabolismo , Riñón/cirugía , Nefrectomía/efectos adversos , Animales , Proteínas Relacionadas con la Autofagia/genética , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/genética , Enfermedades Renales/patología , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Metabolómica , Ratones Endogámicos C57BL , Transducción de Señal
16.
Cells ; 9(7)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32708597

RESUMEN

Podocytes are an integral part of the glomerular filtration barrier, a structure that prevents filtration of large proteins and macromolecules into the urine. Podocyte function is dependent on actin cytoskeleton regulation within the foot processes, structures that link podocytes to the glomerular basement membrane. Actin cytoskeleton dynamics in podocyte foot processes are complex and regulated by multiple proteins and other factors. There are two key signal integration and structural hubs within foot processes that regulate the actin cytoskeleton: the slit diaphragm and focal adhesions. Both modulate actin filament extension as well as foot process mobility. No matter what the initial cause, the final common pathway of podocyte damage is dysregulation of the actin cytoskeleton leading to foot process retraction and proteinuria. Disruption of the actin cytoskeleton can be due to acquired causes or to genetic mutations in key actin regulatory and signaling proteins. Here, we describe the major structural and signaling components that regulate the actin cytoskeleton in podocytes as well as acquired and genetic causes of actin dysregulation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Podocitos/metabolismo , Citoesqueleto de Actina/ultraestructura , Actinas/química , Actinas/genética , Actinas/metabolismo , Animales , Enfermedad/genética , Adhesiones Focales/metabolismo , Humanos , Mutación/genética , Podocitos/ultraestructura
17.
PLoS One ; 15(12): e0230401, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33370294

RESUMEN

Podocytes have been proposed to be antigen presenting cells (APCs). In traditional APCs, the neonatal Fc receptor (FcRn) is required for antigen presentation and global knockout of FcRn protects against glomerulonephritis. Since podocytes express FcRn, we sought to determine whether the absence of podocyte FcRn ameliorates immune-mediated disease. We examined MHCII and costimulatory markers expression in cultured wild type (WT) and FcRn knockout (KO) podocytes. Interferon gamma (IFNγ) induced MHCII expression in both WT and KO podocytes but did not change CD80 expression. Neither WT nor KO expressed CD86 or inducible costimulatory ligand (ICOSL) at baseline or with IFNγ. Using an antigen presentation assay, WT podocytes but not KO treated with immune complexes induced a modest increase in IL-2. Induction of the anti-glomerular basement membrane (anti-GBM) model resulted in a significant decrease in glomerular crescents in podocyte-specific FcRn knockout mouse (podFcRn KO) versus controls but the overall percentage of crescents was low. To examine the effects of the podocyte-specific FcRn knockout in a model with a longer autologous phase, we used the nephrotoxic serum nephritis (NTS) model. We found that the podFcRn KO mice had significantly reduced crescent formation and glomerulosclerosis compared to control mice. This study demonstrates that lack of podocyte FcRn is protective in immune mediated kidney disease that is dependent on an autologous phase. This study also highlights the difference between the anti-GBM model and NTS model of disease.


Asunto(s)
Glomerulonefritis/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Podocitos/metabolismo , Receptores Fc/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Citometría de Flujo , Membrana Basal Glomerular/metabolismo , Glomerulonefritis/genética , Antígenos de Histocompatibilidad Clase I/genética , Ratones , Ratones Noqueados , Receptores Fc/genética
18.
MethodsX ; 7: 101048, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32944514

RESUMEN

Glomerular endothelial cells (GEnC) are a specialized microvascular subset of endothelial cells that, when injured, result in many types of diseases within the kidney. Thus, techniques to study GEnC in a cell culture system are important to investigate mechanisms of GEnC injury. Studies of endothelial cell function in culture have predominately relied on using macrovascular endothelial cells from vascular areas other than the glomerulus. Over the last 15 years, glomerular endothelial cells lines have been created but were isolated by targeting cells expressing CD31. Some studies identified endothelial cells isolated from the microvasculature do not express CD31 and some suggest that CD31+ cells are phenotypically different than endothelial cells found in capillaries. Here we detail our method of isolation, purification, and conditional immortalization of mouse glomerular endothelial cells targeting endothelial cells that do not express CD31.•This method allows for isolation, purification, and conditional immortalization of glomerular endothelial cells for continued passage of GEnCs beyond that of primary cell culture.•This method can be used in genetically modified mice to investigate how a modification of a specific gene or protein affects the glomerular endothelium at the cellular level.

19.
Transpl Immunol ; 58: 101261, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31887408

RESUMEN

Chronic active antibody-mediated rejection is a major cause of allograft failure in kidney transplantation. Microvascular inflammation and transplant glomerulopathy are defining pathologic features of chronic active antibody-mediated rejection and are associated with allograft failure. However, the mechanisms of leukocyte infiltration and glomerular endothelial cell injury remain unclear. We hypothesized MHC class II ligation on glomerular endothelial cells (GEnC) would result in upregulation of adhesion molecules and production of chemoattractants. A model of endothelial cell activation in the presence of antibodies to MHC classes I and II was used to determine the expression of adhesion molecules and chemokines. Murine GEnC were activated with IFNγ, which upregulated gene expression of ß2-microglobulin (MHC class I), ICAM1, VCAM1, CCL2, CCL5, and IL-6. IFNγ stimulation of GEnC increased surface expression of MHC class I, MHC class II, ICAM1, and VCAM1. Incubation with antibodies directed at MHC class I or class II did not further enhance adhesion molecule expression. Multispectral imaging flow cytometry and confocal microscopy demonstrated MHC molecules co-localized with the adhesion molecules ICAM1 and VCAM1 on the GEnC surface. GEnC secretion of chemoattractants, CCL2 and CCL5, was increased by IFNγ stimulation. CCL2 production was further enhanced by incubation with sensitized plasma. Endothelial activation induces de novo expression of MHC class II molecules and increases surface expression of MHC class I, ICAM1 and VCAM1, which are all co-localized together. Maintaining the integrity and functionality of the glomerular endothelium is necessary to ensure survival of the allograft. IFNγ stimulation of GEnC propagates an inflammatory response with production of chemokines and co-localization of MHC and adhesion molecules on the GEnC surface, contributing to endothelial cell function as antigen presenting cells and an active player in allograft injury.


Asunto(s)
Aloinjertos/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Glomérulos Renales/patología , Animales , Presentación de Antígeno , Células Cultivadas , Citometría de Flujo , Isoanticuerpos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Transporte de Proteínas , Regulación hacia Arriba
20.
Am J Physiol Cell Physiol ; 297(6): C1339-46, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19776390

RESUMEN

Parathyroid hormone (PTH) plays a critical role in the regulation of renal phosphorous homeostasis by altering the levels of the sodium-phosphate cotransporter NaPi2a in the brush border membrane (BBM) of renal proximal tubular cells. While details of the molecular events of PTH-induced internalization of NaPi2a are emerging, the precise events governing NaPi2a removal from brush border microvilli in response to PTH remain to be fully determined. Here we use a novel application of total internal reflection fluorescence microscopy to examine how PTH induces movement of NaPi2a out of brush border microvilli in living cells in real time. We show that a dynamic actin cytoskeleton is required for NaPi2a removal from the BBM in response to PTH. In addition, we demonstrate that a myosin motor that has previously been shown to be coregulated with NaPi2a, myosin VI, is necessary for PTH-induced removal of NaPi2a from BBM microvilli.


Asunto(s)
Actinas/metabolismo , Membrana Celular/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Hormona Paratiroidea/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Genes Dominantes , Túbulos Renales Proximales/citología , Microscopía Confocal , Microscopía Fluorescente/métodos , Microvellosidades/efectos de los fármacos , Microvellosidades/metabolismo , Zarigüeyas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA