Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1270980, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38125876

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD; MIM#263200) is a severe, hereditary, hepato-renal fibrocystic disorder that leads to early childhood morbidity and mortality. Typical forms of ARPKD are caused by pathogenic variants in the PKHD1 gene, which encodes the fibrocystin/polyductin (FPC) protein. MYC overexpression has been proposed as a driver of renal cystogenesis, but little is known about MYC expression in recessive PKD. In the current study, we provide the first evidence that MYC is overexpressed in kidneys from ARPKD patients and confirm that MYC is upregulated in cystic kidneys from cpk mutant mice. In contrast, renal MYC expression levels were not altered in several Pkhd1 mutant mice that lack a significant cystic kidney phenotype. We leveraged previous observations that the carboxy-terminus of mouse FPC (FPC-CTD) is proteolytically cleaved through Notch-like processing, translocates to the nucleus, and binds to double stranded DNA, to examine whether the FPC-CTD plays a role in regulating MYC/Myc transcription. Using immunofluorescence, reporter gene assays, and ChIP, we demonstrate that both human and mouse FPC-CTD can localize to the nucleus, bind to the MYC/Myc P1 promoter, and activate MYC/Myc expression. Interestingly, we observed species-specific differences in FPC-CTD intracellular trafficking. Furthermore, our informatic analyses revealed limited sequence identity of FPC-CTD across vertebrate phyla and database queries identified temporal differences in PKHD1/Pkhd1 and CYS1/Cys1 expression patterns in mouse and human kidneys. Given that cystin, the Cys1 gene product, is a negative regulator of Myc transcription, these temporal differences in gene expression could contribute to the relative renoprotection from cystogenesis in Pkhd1-deficient mice. Taken together, our findings provide new mechanistic insights into differential mFPC-CTD and hFPC-CTD regulation of MYC expression in renal epithelial cells, which may illuminate the basis for the phenotypic disparities between human patients with PKHD1 pathogenic variants and Pkhd1-mutant mice.

2.
FASEB J ; 37(7): e23008, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37318790

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD) is caused primarily by mutations in PKHD1, encoding fibrocystin (FPC), but Pkhd1 mutant mice failed to reproduce the human phenotype. In contrast, the renal lesion in congenital polycystic kidney (cpk) mice, with a mutation in Cys1 and cystin protein loss, closely phenocopies ARPKD. Although the nonhomologous mutation diminished the translational relevance of the cpk model, recent identification of patients with CYS1 mutations and ARPKD prompted the investigations described herein. We examined cystin and FPC expression in mouse models (cpk, rescued-cpk (r-cpk), Pkhd1 mutants) and mouse cortical collecting duct (CCD) cell lines (wild type (wt), cpk). We found that cystin deficiency caused FPC loss in both cpk kidneys and CCD cells. FPC levels increased in r-cpk kidneys and siRNA of Cys1 in wt cells reduced FPC. However, FPC deficiency in Pkhd1 mutants did not affect cystin levels. Cystin deficiency and associated FPC loss impacted the architecture of the primary cilium, but not ciliogenesis. No reduction in Pkhd1 mRNA levels in cpk kidneys and CCD cells suggested posttranslational FPC loss. Studies of cellular protein degradation systems suggested selective autophagy as a mechanism. In support of the previously described function of FPC in E3 ubiquitin ligase complexes, we demonstrated reduced polyubiquitination and elevated levels of functional epithelial sodium channel in cpk cells. Therefore, our studies expand the function of cystin in mice to include inhibition of Myc expression via interaction with necdin and maintenance of FPC as functional component of the NEDD4 E3 ligase complexes. Loss of FPC from E3 ligases may alter the cellular proteome, contributing to cystogenesis through multiple, yet to be defined, mechanisms.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo , Humanos , Ratones , Animales , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Proteoma/metabolismo , Receptores de Superficie Celular/metabolismo , Riñón/metabolismo , Factores de Transcripción/metabolismo , Células Epiteliales/metabolismo
4.
Kidney360 ; 1(3): 179-190, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33490963

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease is caused by genetic mutations in PKD1 or PKD2. Macrophages and their associated inflammatory cytokines promote cyst progression; however, transcription factors within macrophages that control cytokine production and cystic disease are unknown. METHODS: In these studies, we used conditional Pkd1 mice to test the hypothesis that macrophage-localized interferon regulatory factor-5 (IRF5), a transcription factor associated with production of cyst-promoting cytokines (TNFα, IL-6), is required for accelerated cyst progression in a unilateral nephrectomy (1K) model. Analyses of quantitative real-time PCR (qRT-PCR) and flow-cytometry data 3 weeks post nephrectomy, a time point before the onset of severe cystogenesis, indicate an accumulation of inflammatory infiltrating and resident macrophages in 1K Pkd1 mice compared with controls. qRT-PCR data from FACS cells at this time demonstrate that macrophages from 1K Pkd1 mice have increased expression of Irf5 compared with controls. To determine the importance of macrophage-localized Irf5 in cyst progression, we injected scrambled or IRF5 antisense oligonucleotide (ASO) in 1K Pkd1 mice and analyzed the effect on macrophage numbers, cytokine production, and renal cystogenesis 6 weeks post nephrectomy. RESULTS: Analyses of qRT-PCR and IRF5 ASO treatment significantly reduced macrophage numbers, Irf5 expression in resident-but not infiltrating-macrophages, and the severity of cystic disease. In addition, IRF5 ASO treatment in 1K Pkd1 mice reduced Il6 expression in resident macrophages, which was correlated with reduced STAT3 phosphorylation and downstream p-STAT3 target gene expression. CONCLUSIONS: These data suggest that Irf5 promotes inflammatory cytokine production in resident macrophages resulting in accelerated cystogenesis.


Asunto(s)
Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Animales , Factores Reguladores del Interferón/genética , Riñón/metabolismo , Macrófagos/metabolismo , Ratones , Enfermedades Renales Poliquísticas/metabolismo , Riñón Poliquístico Autosómico Dominante/genética
5.
Am J Physiol Renal Physiol ; 317(3): F632-F637, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31313950

RESUMEN

Flow-related bending of cilia results in Ca2+ influx through a polycystin-1 (Pkd1) and polycystin-2 (Pkd2) complex, both of which are members of the transient receptor potential (TRP) family (TRPP1 and TRPP2, respectively). Deletion of this complex as well as cilia result in polycystic kidney disease. The Ca2+ influx pathway has been previously characterized in immortalized collecting duct cells without cilia and found to be a 23-pS channel that was a multimere of TRPP2 and TRPV4. The purpose of the present study was to determine if this TRPP2 and TRPV4 multimere exists in vivo. Apical channel activity was measured using the patch-clamp technique from isolated split-open cortical collecting ducts from adult conditional knockout mice with (Ift88flox/flox) or without (Ift88-/-) cilia. Single tubules were isolated for measurements of mRNA for Pkd1, Pkd2, Trpv4, and epithelial Na+ channel subunits. The predominant channel activity from Ift88flox/flox mice was from epithelial Na+ channel [5-pS Na+-selective channels with long mean open times (475.7 ± 83.26 ms) and open probability > 0.2]. With the loss of cilia, the predominant conductance was a 23-pS nonselective cation channel (reversal potential near 0) with a short mean open time (72 ± 17 ms), open probability < 0.08, and a characteristic flickery opening. Loss of cilia increased mRNA levels for Pkd2 and Trpv4 from single isolated cortical collecting ducts. In conclusion, 23-pS channels exist in vivo, and activity of this channel is elevated with loss of cilia, consistent with previous finding of an elevated-unregulated Ca2+-permeable pathway at the apical membrane of collecting duct cells that lack cilia.


Asunto(s)
Cilios/metabolismo , Túbulos Renales Colectores/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Canales Catiónicos TRPP/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Señalización del Calcio , Cilios/patología , Modelos Animales de Enfermedad , Femenino , Túbulos Renales Colectores/patología , Masculino , Potenciales de la Membrana , Ratones Noqueados , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPV/genética , Factores de Tiempo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
6.
Am J Physiol Renal Physiol ; 316(3): F414-F425, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30566001

RESUMEN

Deficiency in polycystin 1 triggers specific changes in energy metabolism. To determine whether defects in other human cystoproteins have similar effects, we studied extracellular acidification and glucose metabolism in human embryonic kidney (HEK-293) cell lines with polycystic kidney and hepatic disease 1 ( PKHD1) and polycystic kidney disease (PKD) 2 ( PKD2) truncating defects along multiple sites of truncating mutations found in patients with autosomal recessive and dominant PKDs. While neither the PKHD1 or PKD2 gene mutations nor their position enhanced cell proliferation rate in our cell line models, truncating mutations in these genes progressively increased overall extracellular acidification over time ( P < 0.001 for PKHD1 and PKD2 mutations). PKHD1 mutations increased nonglycolytic acidification rate (1.19 vs. 1.03, P = 0.002), consistent with an increase in tricarboxylic acid cycle activity or breakdown of intracellular glycogen. In addition, they increased basal and ATP-linked oxygen consumption rates [7.59 vs. 5.42 ( P = 0.015) and 4.55 vs. 2.98 ( P = 0.004)]. The PKHD1 and PKD2 mutations also altered mitochondrial morphology, resembling the effects of polycystin 1 deficiency. Together, these data suggest that defects in major PKD genes trigger changes in mitochondrial energy metabolism. After validation in in vivo models, these initial observations would indicate potential benefits of targeting energy metabolism in the treatment of PKDs.


Asunto(s)
Metabolismo Energético/genética , Glucosa/metabolismo , Proteínas Quinasas/genética , Receptores de Superficie Celular/genética , Proliferación Celular/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Edición Génica , Células HEK293 , Humanos , Mutación , Proteína Quinasa D2 , Proteínas Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo
7.
Free Radic Biol Med ; 102: 260-273, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27913210

RESUMEN

NAFLD is a clinically progressive disease with steatosis, inflammation, endothelial dysfunction and fibrosis being the stages where clinical intervention becomes necessary. Lack of early biomarkers and absence of a FDA approved drug obstructs efforts for effective treatment. NAFLD progression is strongly linked to a balance between liver injury, tissue regeneration and the functioning of endogenous defense mechanisms. The failure of the defense pathways to resist the tissue damage arising from redox stress, one of the "multiple hits" in disease progression, give rise to heightened inflammation and occasional fibrosis. We introduce an endogenous defense mechanism in the liver that is mediated by TRPV4, a transient receptor potential calcium-permeable ion channel that responds to the cytotoxic liver environment and negatively regulates CYP2E1, a cytochrome p450 enzyme. Using Trpv4-/- mice and cultured primary cells, we show that TRPV4 is activated both by damage associated molecular pattern HMGB1 and collagen in diseased Kupffer cells that in turn activate the endothelial NOS (NOS3) to release nitric oxide (NO). The diffusible NO acts in a paracrine fashion in neighboring hepatocytes to deactivate the redox toxicity induced by CYP2E1. We also find that CYP2E1-mediated TRPV4 repression in late stages causes an unrestricted progression of disease. Thus, TRPV4 functions as a sensor of cell stress in the diseased fatty liver and constitutes an endogenous defense molecule, a novel concept with potential for therapeutic approaches against NAFLD, perhaps also against hepatic drug toxicity in general.


Asunto(s)
Citocromo P-450 CYP2E1/genética , Óxido Nítrico Sintasa de Tipo III/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Canales Catiónicos TRPV/genética , Animales , Citocromo P-450 CYP2E1/metabolismo , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Regulación de la Expresión Génica , Proteína HMGB1/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidación-Reducción , Estrés Oxidativo/genética , Canales Catiónicos TRPV/metabolismo , Activación Transcripcional/genética
8.
FASEB J ; 30(1): 370-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26391272

RESUMEN

Activation of the intrarenal renin angiotensin system (RAS) is believed to play an important role in the development of hypertension and cystogenesis in autosomal dominant polycystic kidney disease (ADPKD). Results of clinical studies testing RAS inhibitors in slowing the progression of cystic disease in ADPKD are inconclusive, and we hypothesized that current RAS inhibitors do not adequately suppress intrarenal RAS. For this study, we compared a novel Gen 2 antisense oligonucleotide (ASO) that inhibits angiotensinogen (Agt) synthesis to lisinopril in adult conditional Pkd1 systemic-knockout mice, a model of ADPKD. Six weeks after Pkd1 global gene knockout, the mice were treated with Agt-ASO (66 mg/kg/wk), lisinopril (100 mg/kg/d), PBS (control), or scrambled ASO (66 mg/kg/wk) for 10 wk, followed by tissue collection. Agt ASO resulted in significant reduction in plasma, liver, and kidney Agt, and increased kidney renin compared with control treatments. Kidneys from Agt-ASO-treated mice were not as enlarged and showed reduced cystic volume compared with lisinopril or control treatments. Blood pressure was better controlled with lisinopril than with Agt-ASO. Agt-ASO suppressed cell proliferation in both cystic and noncystic cells compared with lisinopril and control treatments. However, Agt-ASO did not reduce cell proliferation in liver, which indicates that Agt-ASO targets cell signaling pathways that specifically suppresses cystogenesis in the kidney. These data suggest that Agt-ASO effectively attenuates intrarenal RAS and therefore can be a novel and effective agent for treating ADPKD.


Asunto(s)
Angiotensinógeno/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Canales Catiónicos TRPP/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Angiotensinógeno/biosíntesis , Animales , Femenino , Riñón/efectos de los fármacos , Riñón/metabolismo , Lisinopril/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades Renales Poliquísticas/genética , Canales Catiónicos TRPP/genética
9.
Proteomics ; 15(23-24): 4051-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26364553

RESUMEN

Proteomic studies including marine mammals are rare, largely due to the lack of fully sequenced genomes. This has hampered the application of these techniques toward biomarker discovery efforts for monitoring of health and disease in these animals. We conducted a pilot label-free LC-MS/MS study to profile and compare the cerebrospinal fluid from California sea lions with domoic acid toxicosis (DAT) and without DAT. Across 11 samples, a total of 206 proteins were identified (FDR<0.1) using a composite mammalian database. Several peptide identifications were validated using stable isotope labeled peptides. Comparison of spectral counts revealed seven proteins that were elevated in the cerebrospinal fluid from sea lions with DAT: complement C3, complement factor B, dickkopf-3, malate dehydrogenase 1, neuron cell adhesion molecule 1, gelsolin, and neuronal cell adhesion molecule. Immunoblot analysis found reelin to be depressed in the cerebrospinal fluid from California sea lions with DAT. Mice administered domoic acid also had lower hippocampal reelin protein levels suggesting that domoic acid depresses reelin similar to kainic acid. In summary, proteomic analysis of cerebrospinal fluid in marine mammals is a useful tool to characterize the underlying molecular pathology of neurodegenerative disease. All MS data have been deposited in the ProteomeXchange with identifier PXD002105 (http://proteomecentral.proteomexchange.org/dataset/PXD002105).


Asunto(s)
Ácido Kaínico/análogos & derivados , Enfermedades Neurodegenerativas/metabolismo , Proteómica , Leones Marinos/metabolismo , Animales , Ácido Kaínico/líquido cefalorraquídeo , Ácido Kaínico/metabolismo , Proteína Reelina , Leones Marinos/genética
11.
Am J Physiol Renal Physiol ; 309(6): F492-8, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26155843

RESUMEN

Since polycystic kidney disease (PKD) was first noted over 30 years ago to have neoplastic parallels, there has been a resurgent interest in elucidating neoplasia-relevant pathways in PKD. Taking a nontargeted metabolomics approach in the B6(Cg)-Cys1(cpk/)J (cpk) mouse model of recessive PKD, we have now characterized metabolic reprogramming in these tissues, leading to a glutamine-dependent TCA cycle shunt toward total 2-hydroxyglutarate (2-HG) production in cpk compared with B6 wild-type kidney tissue. After confirmation of increased 2-HG expression in immortalized collecting duct cpk cells as well as in human autosomal recessive PKD tissue using targeted analysis, we show that the increase in 2-HG is likely due to glutamine-sourced α-ketoglutarate. In addition, cpk cells require exogenous glutamine for growth such that inhibition of glutaminase-1 decreases cell viability as well as proliferation. This study is a demonstration of the striking parallels between recessive PKD and cancer metabolism. Our data, once confirmed in other PKD models, suggest that future therapeutic approaches targeting this pathway, such as using glutaminase inhibitors, have the potential to open novel treatment options for renal cystic disease.


Asunto(s)
Creatina Quinasa/genética , Glutamina/metabolismo , Glutaratos/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Animales , Células Cultivadas , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Glutaminasa/antagonistas & inhibidores , Humanos , Lactante , Recién Nacido , Masculino , Metabolómica , Ratones , Modelos Genéticos
13.
Physiology (Bethesda) ; 30(3): 195-207, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25933820

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is the most prevalent inherited renal disease, characterized by multiple cysts that can eventually lead to kidney failure. Studies investigating the role of primary cilia and polycystins have significantly advanced our understanding of the pathogenesis of PKD. This review will present clinical and basic aspects of ADPKD, review current concepts of PKD pathogenesis, evaluate potential therapeutic targets, and highlight challenges for future clinical studies.


Asunto(s)
Riñón , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/terapia , Canales Catiónicos TRPP/genética , Animales , Cilios , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Terapia Molecular Dirigida , Fenotipo , Riñón Poliquístico Autosómico Dominante/diagnóstico , Riñón Poliquístico Autosómico Dominante/epidemiología , Riñón Poliquístico Autosómico Dominante/metabolismo , Valor Predictivo de las Pruebas , Factores de Riesgo , Transducción de Señal , Resultado del Tratamiento
14.
Physiol Rep ; 3(5)2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25999403

RESUMEN

The mechanism for early hypertension in polycystic kidney disease (PKD) has not been elucidated. One potential pathway that may contribute to the elevation in blood pressure in PKD is the activation of the intrarenal renin-angiotensin-system (RAS). For example, it has been shown that kidney cyst and cystic fluid contains renin, angiotensin II (AngII), and angiotensinogen (Agt). Numerous studies suggest that ciliary dysfunction plays an important role in PKD pathogenesis. However, it is unknown whether the primary cilium affects the intrarenal RAS in PKD. The purpose of this study was to determine whether loss of cilia or polycystin 1 (PC1) increases intrarenal RAS in mouse model of PKD. Adult Ift88 and Pkd1 conditional floxed allele mice with or without cre were administered tamoxifen to induce global knockout of the gene. Three months after tamoxifen injection, kidney tissues were examined by histology, immunofluorescence, western blot, and mRNA to assess intrarenal RAS components. SV40 immortalized collecting duct cell lines from hypomorphic Ift88 mouse were used to assess intrarenal RAS components in collecting duct cells. Mice without cilia and PC1 demonstrated increased kidney cyst formation, systolic blood pressure, prorenin, and kidney and urinary angiotensinogen levels. Interestingly immunofluorescence study of the kidney revealed that the prorenin receptor was localized to the basolateral membrane of principal cells in cilia (-) but not in cilia (+) kidneys. Collecting duct cAMP responses to AngII administration was greater in cilia (-) vs. cilia (+) cells indicating enhanced intrarenal RAS activity in the absence of cilia. These data suggest that in the absence of cilia or PC1, there is an upregulation of intrarenal RAS components and activity, which may contribute to elevated blood pressure in PKD.

15.
Am J Physiol Renal Physiol ; 309(1): F79-87, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25904703

RESUMEN

In polycystic kidney disease (PKD), the rate of cyst formation and disease progression is highly variable. The lack of predictability in disease progression may be due to additional environmental factors or pathophysiological processes called "third hits." Diabetes is a growing epidemic, and recent studies suggest that PKD patients may be at an increased risk for this disease. We sought to determine if hyperglycemia enhances the initiation and rate of cystogenesis. Tamoxifen was administered to adult Ift88 conditional floxed allele mice to induce cilia loss in the presence of Cre. Subsequent administration of streptozotocin resulted in equivalent hyperglycemia in cilia(+) and cilia(-) mice. Hyperglycemia with loss of cilia increased the rate of cyst formation and cell proliferation. Structural and functional alterations in the kidney, including focal glomerular foot process effacement, interstitial inflammation, formation of primitive renal tubules, polyuria, and increased proteinuria, were also observed in hyperglycemic cilia(-) mice. Gene array analysis indicated enhanced Wnt and epithelial-to-mesenchymal transition signaling in the kidney of hyperglycemic cilia(-) mice. These data show that hyperglycemia, in the absence of cilia, results in renal structural and functional damage and accelerates cystogenesis, suggesting that diabetes is a risk factor in the progression of PKD.


Asunto(s)
Hiperglucemia/complicaciones , Riñón/patología , Enfermedades Renales Poliquísticas/etiología , Animales , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Hemodinámica , Hiperglucemia/patología , Hiperglucemia/fisiopatología , Pruebas de Función Renal , Masculino , Ratones Noqueados , Enfermedades Renales Poliquísticas/patología , Distribución Aleatoria , Proteínas Wnt/metabolismo
17.
J Biol Chem ; 290(11): 6890-902, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25616662

RESUMEN

Microtubule-based centrioles in the centrosome mediate accurate bipolar cell division, spindle orientation, and primary cilia formation. Cellular checkpoints ensure that the centrioles duplicate only once in every cell cycle and achieve precise dimensions, dysregulation of which results in genetic instability and neuro- and ciliopathies. The normal cellular level of centrosomal protein 4.1-associated protein (CPAP), achieved by its degradation at mitosis, is considered as one of the major mechanisms that limits centriole growth at a predetermined length. Here we show that CPAP levels and centriole elongation are regulated by centrobin. Exogenous expression of centrobin causes abnormal elongation of centrioles due to massive accumulation of CPAP in the cell. Conversely, CPAP was undetectable in centrobin-depleted cells, suggesting that it undergoes degradation in the absence of centrobin. Only the reintroduction of full-length centrobin, but not its mutant form that lacks the CPAP binding site, could restore cellular CPAP levels in centrobin-depleted cells, indicating that persistence of CPAP requires its interaction with centrobin. Interestingly, inhibition of the proteasome in centrobin-depleted cells restored the cellular and centriolar CPAP expression, suggesting its ubiquitination and proteasome-mediated degradation when centrobin is absent. Intriguingly, however, centrobin-overexpressing cells also showed proteasome-independent accumulation of ubiquitinated CPAP and abnormal, ubiquitin-positive, elongated centrioles. Overall, our results show that centrobin interacts with ubiquitinated CPAP and prevents its degradation for normal centriole elongation function. Therefore, it appears that loss of centrobin expression destabilizes CPAP and triggers its degradation to restrict the centriole length during biogenesis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Línea Celular , Centriolos/ultraestructura , Eliminación de Gen , Humanos , Proteínas Asociadas a Microtúbulos/análisis , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitinación , Regulación hacia Arriba
18.
Am J Physiol Cell Physiol ; 307(6): C554-60, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25055824

RESUMEN

Epidermal growth factor (EGF) is linked to the pathogenesis of polycystic kidney disease (PKD). We explored signaling pathways activated by EGF in orpk cilia (-) collecting duct cell line derived from a mouse model of PKD (hypomorph of the Tg737/Ift88 gene) with severely stunted cilia, and in a control orpk cilia (+) cell line with normal cilia. RT-PCR demonstrated mRNAs for EGF receptor subunits ErbB1, ErbB2, ErbB3, ErbB4, and mRNAs for Na(+)/H(+) exchangers (NHE), NHE-1, NHE-2, NHE-3, NHE-4, and NHE-5 in both cell lines. EGF stimulated proton efflux in both cell lines. This effect was significantly attenuated by MIA, 5-(n-methyl-N-isobutyl) amiloride, a selective inhibitor of NHE-1 and NHE-2, and orpk cilia (-) cells were more sensitive to MIA than control cells (P < 0.01). EGF significantly induced extracellular signal-regulated kinase (ERK) phosphorylation in both cilia (+) and cilia (-) cells (63.3 and 123.6%, respectively), but the effect was more pronounced in orpk cilia (-) cells (P < 0.01). MIA significantly attenuated EGF-induced ERK phosphorylation only in orpk cilia (-) cells (P < 0.01). EGF increased proliferation of orpk cilia (+) cells and orpk cilia (-) cells, respectively, and MIA at 1-5 µM attenuated EGF-induced proliferation in orpk cilia (-) cells without affecting proliferation of orpk cilia (+) cells. EGF-induced proliferation of both cell lines was significantly decreased by the EGFR tyrosine kinase inhibitor AG1478 and MEK inhibitor PD98059. These results suggest that EGF exerts mitogenic effects in the orpk cilia (-) cells via activation of growth-associated amiloride-sensitive NHEs and ERK.


Asunto(s)
Proliferación Celular , Factor de Crecimiento Epidérmico/metabolismo , Túbulos Renales Colectores/enzimología , Enfermedades Renales Poliquísticas/enzimología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Cilios/enzimología , Cilios/patología , Modelos Animales de Enfermedad , Activación Enzimática , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Isoenzimas , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/patología , Ratones , Ratones Transgénicos , Fosforilación , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/genética , Transfección , Proteínas Supresoras de Tumor/genética
19.
Am J Physiol Renal Physiol ; 307(5): F551-9, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899059

RESUMEN

Polycystic kidney disease (PKD) is a common genetic disorder leading to cyst formation in the kidneys and other organs that ultimately results in kidney failure and death. Currently, there is no therapy for slowing down or stopping the progression of PKD. In this study, we identified the disintegrin metalloenzyme 17 (ADAM17) as a key regulator of cell proliferation in kidney tissues of conditional knockout Ift88(-/-) mice and collecting duct epithelial cells from Ift88°(rpk) mice, animal models of autosomal recessive polycystic kidney disease (ARPKD). Using Western blotting, an enzyme activity assay, and a growth factor-shedding assay in the presence or absence of the specific ADAM17 inhibitor TMI-005, we show that increased expression and activation of ADAM17 in the cystic kidney and in collecting duct epithelial cells originating from the Ift88°(rpk) mice (designated as PKD cells) lead to constitutive shedding of several growth factors, including heparin-binding EGF-like growth factor (HB-EGF), amphiregulin, and transforming growth factor-α (TGF-α). Increased growth factor shedding induces activation of the EGFR/MAPK/ERK pathway and maintains higher cell proliferation rate in PKD cells compared with control cells. PKD cells also displayed increased lactate formation and extracellular acidification indicative of aerobic glycolysis (Warburg effect), which was blocked by ADAM17 inhibition. We propose that ADAM17 is a key promoter of cellular proliferation in PKD cells by activating the EGFR/ERK axis and a proproliferative glycolytic phenotype.


Asunto(s)
Proteínas ADAM/fisiología , Proliferación Celular/fisiología , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Glucólisis/fisiología , Túbulos Renales Colectores/patología , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/efectos de los fármacos , Proteína ADAM17 , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Receptores ErbB/fisiología , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina/fisiología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/fisiopatología , Masculino , Ratones , Ratones Noqueados , Morfolinas/farmacología , Fenotipo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Factor de Crecimiento Transformador alfa/fisiología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
20.
Am J Physiol Renal Physiol ; 307(5): F560-70, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24920756

RESUMEN

Nearly all patients with tuberous sclerosis complex (TSC) develop renal angiomyolipomas, although the tumor cell of origin is unknown. We observed decreased renal angiomyolipoma development in patients with TSC2- polycystic kidney disease 1 deletion syndrome and hypertension that were treated from an early age with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers compared with patients who did not receive this therapy. TSC-associated renal angiomyolipomas expressed ANG II type 1 receptors, platelet-derived growth factor receptor-ß, desmin, α-smooth muscle actin, and VEGF receptor 2 but did not express the adipocyte marker S100 or the endothelial marker CD31. Sera of TSC patients exhibited increased vascular mural cell-secreted peptides, such as VEGF-A, VEGF-D, soluble VEGF receptor 2, and collagen type IV. These findings suggest that angiomyolipomas may arise from renal pericytes. ANG II treatment of angiomyolipoma cells in vitro resulted in an exaggerated intracellular Ca(2+) response and increased proliferation, which were blocked by the ANG II type 2 receptor antagonist valsartan. Blockade of ANG II signaling may have preventative therapeutic potential for angiomyolipomas.


Asunto(s)
Angiomiolipoma/tratamiento farmacológico , Angiomiolipoma/patología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Pericitos/patología , Esclerosis Tuberosa/complicaciones , Angiomiolipoma/fisiopatología , Angiotensina II/fisiología , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Calcio/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Humanos , Técnicas In Vitro , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/fisiopatología , Receptor de Angiotensina Tipo 1/fisiología , Sistema Renina-Angiotensina/fisiología , Transducción de Señal/fisiología , Tetrazoles/farmacología , Tetrazoles/uso terapéutico , Esclerosis Tuberosa/patología , Esclerosis Tuberosa/fisiopatología , Valina/análogos & derivados , Valina/farmacología , Valina/uso terapéutico , Valsartán
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...