Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(12): 5399-5404, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30833403

RESUMEN

Three-dimensional renal tissues that emulate the cellular composition, geometry, and function of native kidney tissue would enable fundamental studies of filtration and reabsorption. Here, we have created 3D vascularized proximal tubule models composed of adjacent conduits that are lined with confluent epithelium and endothelium, embedded in a permeable ECM, and independently addressed using a closed-loop perfusion system to investigate renal reabsorption. Our 3D kidney tissue allows for coculture of proximal tubule epithelium and vascular endothelium that exhibits active reabsorption via tubular-vascular exchange of solutes akin to native kidney tissue. Using this model, both albumin uptake and glucose reabsorption are quantified as a function of time. Epithelium-endothelium cross-talk is further studied by exposing proximal tubule cells to hyperglycemic conditions and monitoring endothelial cell dysfunction. This diseased state can be rescued by administering a glucose transport inhibitor. Our 3D kidney tissue provides a platform for in vitro studies of kidney function, disease modeling, and pharmacology.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Reabsorción Renal , Albúminas/metabolismo , Glucosa/metabolismo , Humanos , Imagenología Tridimensional , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/ultraestructura , Microscopía Electrónica , Modelos Biológicos , Reabsorción Renal/fisiología
2.
Am J Physiol Renal Physiol ; 314(4): F501-F516, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29187371

RESUMEN

The erythropoietin receptor (EpoR) is widely expressed but its renoprotective action is unexplored. To examine the role of EpoR in vivo in the kidney, we induced acute kidney injury (AKI) by ischemia-reperfusion in mice with different EpoR bioactivities in the kidney. EpoR bioactivity was reduced by knockin of wild-type human EpoR, which is hypofunctional relative to murine EpoR, and a renal tubule-specific EpoR knockout. These mice had lower EPO/EpoR activity and lower autophagy flux in renal tubules. Upon AKI induction, they exhibited worse renal function and structural damage, more apoptosis at the acute stage (<7 days), and slower recovery with more tubulointerstitial fibrosis at the subacute stage (14 days). In contrast, mice with hyperactive EpoR signaling from knockin of a constitutively active human EpoR had higher autophagic flux, milder kidney damage, and better renal function at the acute stage but, surprisingly, worse tubulointerstitial fibrosis and renal function at the subacute stage. Either excess or deficient EpoR activity in the kidney was associated with abnormal peritubular capillaries and tubular hypoxia, creating a "U-shaped" relationship. The direct effects of EpoR on tubular cells were confirmed in vitro by a hydrogen peroxide model using primary cultured proximal tubule cells with different EpoR activities. In summary, normal erythropoietin (EPO)/EpoR signaling in renal tubules provides defense against renal tubular injury maintains the autophagy-apoptosis balance and peritubular capillary integrity. High and low EPO/EpoR bioactivities both lead to vascular defect, and high EpoR activity overides the tubular protective effects in AKI recovery.


Asunto(s)
Lesión Renal Aguda/metabolismo , Capilares/metabolismo , Eritropoyetina/metabolismo , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/metabolismo , Neovascularización Fisiológica , Receptores de Eritropoyetina/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Apoptosis , Autofagia , Capilares/patología , Capilares/fisiopatología , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Humanos , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Ratones de la Cepa 129 , Ratones Transgénicos , Receptores de Eritropoyetina/deficiencia , Receptores de Eritropoyetina/genética , Transducción de Señal
3.
Kidney Int ; 94(3): 524-535, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30045814

RESUMEN

Multiple large clinical trials have shown that sodium-glucose cotransporter (SGLT) 2 inhibitors reduce the risk of renal events. However, the mechanism responsible for this outcome remains unknown. Here we investigated the effects of the SGLT2 inhibitor luseogliflozin on the development of renal fibrosis after renal ischemia/reperfusion injury in non-diabetic mice. Luseogliflozin significantly suppressed development of renal fibrosis, prevented peritubular capillary congestion/hemorrhage, attenuated CD31-positive cell loss, suppressed hypoxia, and increased vascular endothelial growth factor (VEGF)-A expression in the kidney after ischemia/reperfusion injury. Luseogliflozin failed to induce the above-mentioned protection in animals co-treated with sunitinib, a VEGF receptor inhibitor. Additionally, luseogliflozin reduced glucose uptake and increased VEGF-A expression in the kidneys of glucose transporter 2 (GLUT2)-downregulated mice following ischemia/reperfusion and in GLUT2-knock-down cells compared with those in normal controls. Withdrawal of glucose from cultured medium, to halt glucose uptake, remarkably increased VEGF-A expression and reversed the luseogliflozin-induced increase in VEGF-A expression in the proximal tubular cells. Thus, luseogliflozin prevented endothelial rarefaction and subsequent renal fibrosis after renal ischemia/reperfusion injury through a VEGF-dependent pathway induced by the dysfunction of proximal tubular glucose uptake in tubules with injury-induced GLUT2 downregulation.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Túbulos Renales Proximales/patología , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Inhibidores de la Angiogénesis/farmacología , Animales , Glucemia/metabolismo , Capilares/efectos de los fármacos , Capilares/metabolismo , Capilares/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Fibrosis , Técnicas de Silenciamiento del Gen , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Humanos , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Daño por Reperfusión/complicaciones , Daño por Reperfusión/patología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Sorbitol/análogos & derivados , Sorbitol/farmacología , Sorbitol/uso terapéutico , Sunitinib/farmacología , Resultado del Tratamiento
4.
Kidney Int ; 87(6): 1125-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25651362

RESUMEN

MicroRNAs, activated by the enzyme Dicer1, control post-transcriptional gene expression. Dicer1 has important roles in the epithelium during nephrogenesis, but its function in stromal cells during kidney development is unknown. To study this, we inactivated Dicer1 in renal stromal cells. This resulted in hypoplastic kidneys, abnormal differentiation of the nephron tubule and vasculature, and perinatal mortality. In mutant kidneys, genes involved in stromal cell migration and activation were suppressed as were those involved in epithelial and endothelial differentiation and maturation. Consistently, polarity of the proximal tubule was incorrect, distal tubule differentiation was diminished, and elongation of Henle's loop attenuated resulting in lack of inner medulla and papilla in stroma-specific Dicer1 mutants. Glomerular maturation and capillary loop formation were abnormal, whereas peritubular capillaries, with enhanced branching and increased diameter, formed later. In Dicer1-null renal stromal cells, expression of factors associated with migration, proliferation, and morphogenic functions including α-smooth muscle actin, integrin-α8, -ß1, and the WNT pathway transcriptional regulator LEF1 were reduced. Dicer1 mutation in stroma led to loss of expression of distinct microRNAs. Of these, miR-214, -199a-5p, and -199a-3p regulate stromal cell functions ex vivo, including WNT pathway activation, migration, and proliferation. Thus, Dicer1 activity in the renal stromal compartment regulates critical stromal cell functions that, in turn, regulate differentiation of the nephron and vasculature during nephrogenesis.


Asunto(s)
Diferenciación Celular/genética , ARN Helicasas DEAD-box/fisiología , Neovascularización Fisiológica/genética , Nefronas/embriología , Ribonucleasa III/fisiología , Actinas/metabolismo , Animales , Capilares/embriología , Movimiento Celular/genética , Proliferación Celular/genética , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Femenino , Expresión Génica , Cadenas alfa de Integrinas/metabolismo , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/citología , Glomérulos Renales/embriología , Túbulos Renales/irrigación sanguínea , Túbulos Renales/citología , Túbulos Renales/embriología , Túbulos Renales Distales/irrigación sanguínea , Túbulos Renales Distales/citología , Túbulos Renales Distales/embriología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/embriología , Asa de la Nefrona/irrigación sanguínea , Asa de la Nefrona/citología , Asa de la Nefrona/embriología , Ratones , MicroARNs/genética , Nefronas/anomalías , Nefronas/citología , Organogénesis/genética , Podocitos/fisiología , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Células del Estroma/fisiología , Transcriptoma , Uréter/anomalías , Vía de Señalización Wnt/genética
5.
Am J Physiol Endocrinol Metab ; 304(12): E1365-78, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23632630

RESUMEN

Matrix metalloproteinase-9 (MMP-9) causes adverse remodeling, whereas hydrogen sulfide (H2S) rescues organs in vascular diseases. The involvement of MMP-9 and H2S in diabetic renovascular remodeling is, however, not well characterized. We determined whether MMP-9 regulates H2S generation and whether H2S modulates connexin through N-methyl-d-aspartate receptor (NMDA-R)-mediated pathway in the diabetic kidney. Wild-type (WT, C57BL/6J), diabetic (Akita, C57BL/6J-Ins2(Akita)), MMP-9(-/-) (M9KO), double knockout (DKO) of Akita/MMP-9(-/-) mice and in vitro cell culture were used in our study. Hyperglycemic Akita mice exhibited increased level of MMP-9 and decreased production of H2S. H2S-synthesizing enzymes cystathionine-ß-synthase and cystathionine-γ-lyase were also diminished. In addition, increased expressions of NMDA-R1 and connexin-40 and -43 were observed in diabetic kidney. As expected, MMP-9 mRNA was not detected in M9KO kidneys. However, very thin protein expression and activity were detected. No other changes were noticed in M9KO kidney. In DKO mice, all the above molecules showed a trend toward baseline despite hyperglycemia. In vitro, glomerular endothelial cells treated with high glucose showed induction of MMP-9, attenuated H2S production, NMDA-R1 induction, and dysregulated conexin-40 and -43 expressions. Silencing MMP-9 by siRNA or inhibition of NMDA-R1 by MK801 or H2S treatment preserved connexin-40 and -43. We conclude that in diabetic renovascular remodeling MMP-9 plays a major role and that H2S has therapeutic potential to prevent adverse diabetic renal remodeling.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Sulfuro de Hidrógeno/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Células Cultivadas , Nefropatías Diabéticas/genética , Maleato de Dizocilpina/farmacología , Células Endoteliales/citología , Antagonistas de Aminoácidos Excitadores/farmacología , Glucosuria Renal/genética , Glucosuria Renal/metabolismo , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/citología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/citología , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
6.
Cells ; 11(24)2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36552750

RESUMEN

Although recent studies have reported that long non-coding RNA (lncRNA) is involved in the development of ischemic acute kidney injury (AKI), the exact function and regulatory mechanism of lncRNAs in ischemic AKI remain largely unknown. Herein, we found that ischemic injury promoted the expression of lncRNA 148400 in mouse proximal tubule-derived cell line (BUMPT) and C57BL/6J mice. Furthermore, the lncRNA148400 mediates ischemic injury-induced apoptosis of BUMPT cells. Mechanistically, lncRNA 148400 sponged miR-10b-3p to promote apoptosis via GRK4 upregulation. Finally, knockdown of lncRNA 148400 alleviated the I/R-induced deterioration of renal function, renal tubular injury, and cell apoptosis. In addition, cleaved caspase-3 is increased via targeting the miR-10b-3p/GRK4 axis. Collectively, these results showed that lncRNA 148400/miR-10b-3p/GRK4 axis mediated the development of ischemic AKI.


Asunto(s)
Lesión Renal Aguda , Células Epiteliales , Túbulos Renales Proximales , ARN Largo no Codificante , Animales , Ratones , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Apoptosis/genética , Apoptosis/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Isquemia/genética , Isquemia/metabolismo
7.
Am J Physiol Renal Physiol ; 301(2): F387-95, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21561997

RESUMEN

Renal proximal tubular epithelium can regenerate after various insults. To examine whether the tubular repair process is regulated by surrounding peritubular capillaries, we established an in vitro human tubulogenesis model that mimics in vivo tubular regeneration after injury. In this model, HGF, a potent renotropic factor, dose dependently induced tubular structures in human renal proximal tubular epithelial cells cultured in gels. Consistent with regenerating tubular cells after injury, HGF-induced tubular structures expressed a developmental gene, Pax-2, and a mesenchymal marker, vimentin, and formed a lumen with aquaporin-1 expression. Electron microscopic analysis showed the presence of microvilli on the apical site of the lumen, suggesting that these structures are morphologically equivalent to renal tubules in vivo. When cocultured with human umbilical vein endothelial cells (HUVEC), HGF-induced tubular formation was significantly enhanced. This could not be reproduced by the addition of VEGF, basic FGF, or PDGF. Protein array revealed that HUVEC produced various matrix metalloproteinases (MMPs). The stimulatory effects of coculture with HUVEC or HUVEC-derived conditional medium were almost completely abolished by addition of the tissue inhibitor of metalloproteinase (TIMP)-1 or TIMP-2. These data suggest that endothelial cell-derived factors including MMPs play a critical role in tubulogenesis and imply a potential role of peritubular capillary endothelium as a source of factor(s) required for tubular recovery after injury.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Túbulos Renales Proximales/fisiología , Regeneración , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Isquemia/patología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratas , Ratas Wistar
8.
Am J Physiol Renal Physiol ; 300(2): F488-98, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21048020

RESUMEN

Peritubular vascular changes and hypoxia after glomerular injury may explain subsequent tubulointerstitial injury and fibrosis. Several studies suggested that the expected tubulointerstitial angiogenic response is actively suppressed in this setting. The mechanism of this aberrant response has not been clearly identified. We used a common model of glomerular injury in rats to assess vascular changes and to identify potential factors associated with this aberrant response. Anti-Thy1.1 antibody administration (1 or 4 weekly doses) led to a dose-dependent renal damage characterized by elevated urea and tubulointerstitial fibrosis as assessed by Picro-Sirius Red staining. We quantified peritubular capillaries using CD31 and CD34 immunohistochemistry and showed that tubular angiogenic dysregulation was associated with peritubular capillary rarefaction. Using laser capture microdissection, we demonstrated an early induction of fibrogenic and angiogenic factors in the glomeruli and a subsequent dysregulated angiogenic response in the tubulointerstitial compartment. Proximal tubules of anti-Thy1.1-treated animals had increased pigment epithelial-derived factor (PEDF) expression by immunohistochemistry. Protein taken by laser capture microdissection also showed that PEDF was upregulated. Temporally associated with PEDF expression was a transient downregulation of tubular hypoxia-inducible factor (HIF)1α. In a human proximal tubular cell culture, we show that PEDF downregulates HIF1α protein and gene expression in cells exposed to 1% oxygen. In anti-Thy1.1 glomerulonephritis, there is aberrent tubular angiogenesis associated with glomerular injury and tubulointersititial fibrosis. We showed that PEDF may be involved by downregulating HIF1α. Further work is needed to elucidate the mechanism of PEDF upregulation and action in the tubules.


Asunto(s)
Glomerulonefritis/fisiopatología , Túbulos Renales Proximales/fisiopatología , Neovascularización Patológica/fisiopatología , Animales , Antígenos CD34/análisis , Antígenos CD34/inmunología , Línea Celular , Regulación hacia Abajo , Proteínas del Ojo/metabolismo , Femenino , Glomerulonefritis/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fallo Renal Crónico/fisiopatología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/patología , Factores de Crecimiento Nervioso/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Ratas , Ratas Sprague-Dawley , Serpinas/metabolismo , Antígenos Thy-1/inmunología , Regulación hacia Arriba , Urea/sangre , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Kidney Int ; 80(2): 181-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21389969

RESUMEN

An acute increase in the international normalized ratio (INR; a comparison of prothrombin time to monitor the effects of warfarin) over 3 in patients with chronic kidney disease (CKD) is often associated with an unexplained acute increase in serum creatinine (SC) and an accelerated progression of CKD. Kidney biopsy in a subset of these patients showed obstruction of the renal tubule by red blood cell casts, and this appears to be the dominant mechanism of the acute kidney injury. We termed this warfarin-related nephropathy (WRN), and previously reported cases of WRN only in patients with CKD. We now assess whether this occurs in patients without CKD, its risk factors, and consequences. In 15,258 patients who initiated warfarin therapy during a 5-year period, 4006 had an INR over 3 and SC measured at the same time; however, the large data set precluded individual patient clinical assessment. A presumptive diagnosis of WRN was made if the SC increased by over 0.3 mg/dl within 1 week after the INR exceeded 3 with no record of hemorrhage. WRN occurred in 20.5% of the entire cohort, 33.0% of the CKD cohort, and 16.5% of the no-CKD cohort. Other risk factors included age, diabetes mellitus, hypertension, and cardiovascular disease. The 1-year mortality was 31.1% with compared with 18.9% without WRN, an increased risk of 65%. Thus, WRN may be a common complication of warfarin therapy in high-risk patients and CKD doubles this risk. The mechanisms of these risks are unclear.


Asunto(s)
Enfermedades Renales/inducido químicamente , Enfermedades Renales/epidemiología , Warfarina/efectos adversos , Biopsia , Enfermedad Crónica , Agregación Eritrocitaria , Humanos , Relación Normalizada Internacional , Enfermedades Renales/mortalidad , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/patología , Mortalidad , Factores de Riesgo
10.
Kidney Int ; 79(11): 1163-5, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21566638

RESUMEN

Hexokinases have emerged as novel mediators of the antiapoptotic effects of growth factors in a wide variety of cells. These effects have been attributed to highly regulated direct physical and functional interactions with mitochondria. The demonstration that mitochondrial hexokinases can prevent apoptogenic 'Bax attack' in proximal tubule cells suggests a need to reexamine the specific contributions of hexokinases and glucose metabolism in this nephron segment and elsewhere within the kidney.


Asunto(s)
Células Epiteliales/enzimología , Hexoquinasa/metabolismo , Enfermedades Renales/enzimología , Túbulos Renales Proximales/enzimología , Membranas Mitocondriales/enzimología , Daño por Reperfusión/enzimología , Animales , Apoptosis , Supervivencia Celular , Células Epiteliales/patología , Hexoquinasa/genética , Humanos , Enfermedades Renales/patología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/patología , Membranas Mitocondriales/patología , Transporte de Proteínas , Daño por Reperfusión/patología , Transducción de Señal , Estrés Fisiológico , Proteína X Asociada a bcl-2/metabolismo
11.
Kidney Int ; 79(11): 1207-16, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21430642

RESUMEN

Hexokinase (HK), the rate-limiting enzyme in glycolysis, controls cell survival by promoting metabolism and/or inhibiting apoptosis. Since HK isoforms I and II have mitochondrial targeting sequences, we attempted to separate the protective effects of HK on cell metabolism from those on apoptosis. We exposed renal epithelial cells to metabolic stress causing ATP depletion in the absence of glucose and found that this activated glycogen synthase kinase 3ß (GSK3ß) and Bax caused mitochondrial membrane injury and apoptosis. ATP depletion led to a progressive HK II dissociation from mitochondria, released mitochondrial apoptosis inducing factor and cytochrome c into the cytosol, activated caspase-3, and reduced cell survival. Compared with control, adenoviral-mediated HK I or II overexpression improved cell survival following stress, but did not prevent GSK3ß or Bax activation, improve ATP content, or reduce mitochondrial fragmentation. HK I or HK II overexpression increased mitochondria-associated isoform-specific HK content, and decreased mitochondrial membrane injury and apoptosis after stress. In vivo, HK II localized exclusively to the proximal tubule. Ischemia reduced total renal HK II content and dissociated HK II from proximal tubule mitochondria. In cells overexpressing HK II, Bax and HK II did not interact before or after stress. While the mechanism by which HK antagonizes Bax-mediated apoptosis is unresolved by these studies, one possible scenario is that the two proteins compete for a common binding site on the outer mitochondrial membrane.


Asunto(s)
Células Epiteliales/enzimología , Hexoquinasa/metabolismo , Enfermedades Renales/enzimología , Túbulos Renales Proximales/enzimología , Membranas Mitocondriales/enzimología , Daño por Reperfusión/enzimología , Estrés Fisiológico , Proteína X Asociada a bcl-2/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Supervivencia Celular , Células Cultivadas , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Glucosa/deficiencia , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hexoquinasa/genética , Enfermedades Renales/patología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/patología , Ratones , Membranas Mitocondriales/patología , Zarigüeyas , Transporte de Proteínas , Daño por Reperfusión/patología , Transducción de Señal , Factores de Tiempo , Transfección
12.
Stem Cell Reports ; 16(1): 134-148, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33306987

RESUMEN

MicroRNAs (miRNAs) are gene expression regulators and they have been implicated in acquired kidney diseases and in renal development, mostly through animal studies. We hypothesized that the miR-199a/214 cluster regulates human kidney development. We detected its expression in human embryonic kidneys by in situ hybridization. To mechanistically study the cluster, we used 2D and 3D human embryonic stem cell (hESC) models of kidney development. After confirming expression in each model, we inhibited the miRNAs using lentivirally transduced miRNA sponges. This reduced the WT1+ metanephric mesenchyme domain in 2D cultures. Sponges did not prevent the formation of 3D kidney-like organoids. These organoids, however, contained dysmorphic glomeruli, downregulated WT1, aberrant proximal tubules, and increased interstitial capillaries. Thus, the miR-199a/214 cluster fine-tunes differentiation of both metanephric mesenchymal-derived nephrons and kidney endothelia. While clinical implications require further study, it is noted that patients with heterozygous deletions encompassing this miRNA locus can have malformed kidneys.


Asunto(s)
Diferenciación Celular , MicroARNs/metabolismo , Antagomirs/metabolismo , Capilares/patología , Técnicas de Cultivo de Célula , Regulación hacia Abajo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Modelos Biológicos , Neovascularización Fisiológica , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
13.
Sci Rep ; 11(1): 16270, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381142

RESUMEN

mTOR inhibitors offer advantages after kidney transplantation including antiviral and antitumor activity besides facilitating low calcineurin inhibitor exposure to reduce nephrotoxicity. Concerns about adverse effects due to antiproliferative and antiangiogenic properties have limited their clinical use particularly early after transplantation. Interference with vascular endothelial growth factor (VEGF)-A, important for physiologic functioning of renal endothelial cells and tubular epithelium, has been implicated in detrimental renal effects of mTOR inhibitors. Low doses of Rapamycin (loading dose 3 mg/kg bodyweight, daily doses 1.5 mg/kg bodyweight) were administered in an allogenic rat kidney transplantation model resulting in a mean through concentration of 4.30 ng/mL. Glomerular and peritubular capillaries, tubular cell proliferation, or functional recovery from preservation/reperfusion injury were not compromised in comparison to vehicle treated animals. VEGF-A, VEGF receptor 2, and the co-receptor Neuropilin-1 were upregulated by Rapamycin within 7 days. Rat proximal tubular cells (RPTC) responded in vitro to hypoxia with increased VEGF-A and VEGF-R1 expression that was not suppressed by Rapamycin at therapeutic concentrations. Rapamycin did not impair proliferation of RPTC under hypoxic conditions. Low-dose Rapamycin early posttransplant does not negatively influence the VEGF network crucial for recovery from preservation/reperfusion injury. Enhancement of VEGF signaling peritransplant holds potential to further improve outcomes.


Asunto(s)
Trasplante de Riñón , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/fisiología , Resultados Negativos , Regeneración/efectos de los fármacos , Sirolimus/efectos adversos , Animales , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Túbulos Renales Proximales/citología , Masculino , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Daño por Reperfusión/genética , Sirolimus/administración & dosificación , Sirolimus/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología
14.
J Am Soc Nephrol ; 20(7): 1544-55, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19443641

RESUMEN

Acute renal ischemia elicits an inflammatory response that may exacerbate acute kidney injury, but the regulation of the initial signals that recruit leukocytes is not well understood. Here, we found that IFN regulatory factor 1 (IRF-1) was a critical, early proinflammatory signal released during ischemic injury in vitro and in vivo. Within 15 min of reperfusion, proximal tubular cells of the S3 segment produced IRF-1, which is a transcription factor that activates proinflammatory genes. Transgenic knockout of IRF-1 ameliorated the impairment of renal function, morphologic injury, and inflammation after acute ischemia. Bone marrow chimera experiments determined that maximal ischemic injury required IRF-1 expression by both leukocytes and radioresistant renal cells, the latter identified as S3 proximal tubule cells in the outer medulla by in situ hybridization and immunohistochemistry. In vitro, reactive oxygen species, generated during ischemia/reperfusion injury, stimulated expression of IRF-1 in an S3 proximal tubular cell line. Taken together, these data suggest that IRF-1 gene activation by reactive oxygen species is an early signal that promotes inflammation after ischemic renal injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Inflamación/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Isquemia/metabolismo , Lesión Renal Aguda/patología , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Células Cultivadas , Modelos Animales de Enfermedad , Inflamación/patología , Factor 1 Regulador del Interferón/genética , Isquemia/patología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/fisiología
15.
Apoptosis ; 14(10): 1176-89, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19685188

RESUMEN

Reactive oxygen species (ROS) contribute significantly to apoptosis in renal ischemia-reperfusion (IR) injury, however the exact mechanisms are not well understood. We used novel lentiviral vectors to over-express superoxide dismutase 1 (SOD1) in proximal tubular epithelial (LLC-PK(1)) cells and determined effects of SOD1 following ATP depletion-recovery, used as a model to simulate renal IR. SOD1 over-expression partially protected against cytotoxicity (P < 0.001) and decreased superoxide (O(2) (*-)) in ATP depleted cells. The ATP depletion-mediated increase in nuclear fragmentation, an index of apoptosis and activation of caspase-3 was also partially blocked by SOD1 (P < 0.05). However, SOD1 over-expression was insufficient to completely attenuate caspase-3, indicating that ROS other than cytoplasmic O(2) (*-) are involved in ATP depletion mediated injury. To test the contribution of hydrogen peroxide, a subset of enhanced green fluorescent protein (EGFP) and SOD1 (serum free and injured) cells were treated with polyethylene glycol-catalase (PEG-catalase). As expected there was 50% reduction in cytotoxicity and caspase-3 in SOD1 cells compared to EGFP cells; catalase treatment decreased both indices by an additional 28% following ATP depletion. To test the role of mitochondrial derived superoxide, we also treated a subset of LLC-PK(1) cells with the mitochondrial antioxidant, MitoTEMPO. Treatment with MitoTEMPO also decreased ATP depletion induced cytotoxicity in LLC-PK(1) cells in a dose dependant manner. These studies indicate that both SOD1 dependent and independent pathways are integral in protection against ATP depletion-recovery mediated cytotoxicity and apoptosis, however more studies are needed to delineate the signaling mechanisms involved.


Asunto(s)
Apoptosis , Células Epiteliales/enzimología , Isquemia/enzimología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/citología , Superóxido Dismutasa/metabolismo , Adenosina Trifosfato/deficiencia , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Catalasa/metabolismo , Fragmentación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Células LLC-PK1 , Lentivirus/genética , Piperidinas/farmacología , Reproducibilidad de los Resultados , Superóxido Dismutasa-1 , Superóxidos/metabolismo , Porcinos , Factores de Tiempo
16.
BJU Int ; 103(2): 254-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18782307

RESUMEN

OBJECTIVE: To examine whether hypoxia (one of the many components of ischaemic preconditioning) can induce a protective response in culture renal tubular cells, and thus determine if non-lethal periods of hypoxia could confer protection against apoptotic injury to human proximal tubular cells during cold storage and subsequent cytotoxic insult, and establish the cellular mechanisms by which this protection is induced. MATERIALS AND METHODS: Human proximal tubular cells (HK-2) were pre-incubated for 24 h in normoxic or hypoxic conditions and then incubated at 4 degrees C for 6 h to mimic cold storage, before being returned to normal conditions and exposed to varying concentrations of cyclosporine A (CSA). Cell viability and apoptosis were measured using propidium iodide staining and flow cytometry. The expression of heat-shock protein (HSP)-70 was determined by Western blotting. RESULTS: Hypoxia had no effect on cell viability or apoptosis. Pre-exposure of cells to hypoxia significantly protected against CSA-induced damage even after a period of cold storage. Western blotting analysis showed that hypoxia up-regulated the anti-apoptotic protein HSP-70. HK-2 cells over-expressing HSP-70 mimicked hypoxia preconditioning, in that they were protected during cold storage and CSA-induced apoptosis. CONCLUSION: Exposure of renal tubular cells to a sequential model of cold storage, reperfusion and incubation with CSA resulted in apoptotic cell death. Preconditioning these cells with hypoxia induced a protective response and up-regulation of the anti-apoptotic protein HSP-70. There was a similar response in non-preconditioned cells over-expressing HSP-70. Further understanding of the cellular changes occurring during this period of preconditioning will allow the development of more targeted, clinically relevant methods of preconditioning in renal transplantation.


Asunto(s)
Hipoxia de la Célula/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Precondicionamiento Isquémico , Túbulos Renales Proximales/irrigación sanguínea , Conservación de Tejido/métodos , Western Blotting , Supervivencia Celular , Ciclosporina/efectos adversos , Citometría de Flujo , Humanos , Regulación hacia Arriba
17.
Nephron Physiol ; 113(4): p23-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19713718

RESUMEN

BACKGROUND/AIMS: Disrupting the enzyme Cyp4a14 in mice leads to hypertension, which is more severe in the male mice and appears to be due to androgen excess. Because the Cyp4a14 enzyme is located in the proximal tubule of the kidney, we hypothesized that there could be dysregulation of transport in this segment that could contribute to the hypertension. METHODS: Wild-type (SV/129) mice and mice that had targeted disruption of the Cyp4a14 gene were studied. Proximal convoluted tubules (PCT) from knockout and wild-type mice were dissected and perfused in vitrofor measurement of volume absorption (J(V)). Expression of the sodium-hydrogen exchanger 3 (NHE3), the predominant transporter responsible for sodium transport in this segment, was measured by immunoblot. Renal vascular (afferent arteriole) responses to angiotensin and endothelin were also measured. RESULTS: PCT volume absorption was elevated in tubules from the Cyp4a14 knockout mice as compared to the wild-type mice. Brush border membrane NHE3 expression was almost 2-fold higher in Cyp4a14 knockout mice than in wild-type mice. No difference was found in the afferent arteriolar response. CONCLUSION: Thus, hypertension in the Cyp4a14 knockout mice appears to be driven by excessive fluid reabsorption in the proximal tubule, which is secondary to overexpression of NHE3.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Hipertensión/metabolismo , Túbulos Renales Proximales/metabolismo , Andrógenos/metabolismo , Animales , Arteriolas/fisiología , Transporte Biológico/fisiología , Presión Sanguínea/fisiología , Sistema Enzimático del Citocromo P-450/genética , Familia 4 del Citocromo P450 , Modelos Animales de Enfermedad , Hipertensión/genética , Túbulos Renales Proximales/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Sodio/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo
18.
Sci Rep ; 9(1): 7883, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-31133638

RESUMEN

In acute kidney injury (AKI), the S3 segment of the proximal tubule is particularly damaged, as it is most vulnerable to ischemia. However, this region is also involved in renal tubular regeneration. To deeply understand the mechanism of the repair process after ischemic injury in AKI, we focused on glial cells missing 1 (Gcm1), which is one of the genes expressed in the S3 segment. Gcm1 is essential for the development of the placenta, and Gcm1 knockout (KO) is embryonically lethal. Thus, the function of Gcm1 in the kidney has not been analyzed yet. We analyzed the function of Gcm1 in the kidney by specifically knocking out Gcm1 in the kidney. We created an ischemia-reperfusion injury (IRI) model to observe the repair process after AKI. We found that Gcm1 expression was transiently increased during the recovery phase of IRI. In Gcm1 conditional KO mice, during the recovery phase of IRI, tubular cell proliferation reduced and transforming growth factor-ß1 expression was downregulated resulting in a reduction in fibrosis. In vitro, Gcm1 overexpression promoted cell proliferation and upregulated TGF-ß1 expression. These findings indicate that Gcm1 is involved in the mechanisms of fibrosis and cell proliferation after ischemic injury of the kidney.


Asunto(s)
Lesión Renal Aguda/patología , Proteínas de Unión al ADN/metabolismo , Túbulos Renales Proximales/patología , Regeneración/fisiología , Daño por Reperfusión/patología , Factores de Transcripción/metabolismo , Lesión Renal Aguda/etiología , Animales , Proliferación Celular/fisiología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Fibrosis , Humanos , Túbulos Renales Proximales/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión/complicaciones , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta1/metabolismo
20.
Nephron Exp Nephrol ; 108(1): e11-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18097150

RESUMEN

BACKGROUND/AIMS: Apoptosis has been implicated in renal ischemic injury, the regulating mechanism of which is still unclear. Signal transducers and activators of transcription (STAT) participate in inflammation, apoptosis, and tumorigenesis. In the in vitro model of renal ischemic injury, we explored the role of the STAT3, a major component of the STAT family, in apoptosis of human proximal tubular epithelial cell (HKC) induced by ATP depletion/recovery. METHODS: Apoptosis of HKC was induced in an in vitro model of acute renal failure. STAT3 activation was analyzed by Western blotting. RNA interference was used to knock down STAT3 expression. The effect of STAT3 knockdown or STAT3C overexpression on apoptosis was assessed by annexin V binding and propidium iodide uptake. RESULTS: STAT3 was phosphorylated during the course of ATP depletion-induced HKC cell apoptosis. STAT3 knockdown suppressed STAT3 phosphorylation, and promoted apoptosis of HKC subjected to ATP depletion/recovery, while STAT3C overexpression conferred resistance of HKC to ATP depletion-induced apoptosis. CONCLUSION: Our results demonstrated that STAT3 mediates resistance to ATP depletion-induced apoptosis of HKC, which may be a potential target in treatment of renal ischemic injury.


Asunto(s)
Adenosina Trifosfato/metabolismo , Apoptosis/fisiología , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Factor de Transcripción STAT3/fisiología , Adenosina Trifosfato/deficiencia , Diferenciación Celular/fisiología , Células Cultivadas , Células Epiteliales/citología , Humanos , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Túbulos Renales Proximales/irrigación sanguínea , Túbulos Renales Proximales/citología , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA