Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Biol ; 500: 1-9, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37209936

RESUMEN

ARL13B is a small GTPase enriched in cilia. Deletion of Arl13b in mouse kidney results in renal cysts and an associated absence of primary cilia. Similarly, ablation of cilia leads to kidney cysts. To investigate whether ARL13B functions from within cilia to direct kidney development, we examined kidneys of mice expressing an engineered cilia-excluded ARL13B variant, ARL13BV358A. These mice retained renal cilia and developed cystic kidneys. Because ARL13B functions as a guanine nucleotide exchange factor (GEF) for ARL3, we examined kidneys of mice expressing an ARL13B variant that lacks ARL3 GEF activity, ARL13BR79Q. We found normal kidney development with no evidence of cysts in these mice. Taken together, our results show that ARL13B functions within cilia to inhibit renal cystogenesis during mouse development, and that this function does not depend on its role as a GEF for ARL3.


Asunto(s)
Enfermedades Renales Quísticas , Riñón , Animales , Ratones , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Cilios/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Riñón/metabolismo , Enfermedades Renales Quísticas/genética
2.
bioRxiv ; 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-36798281

RESUMEN

ARL13B is a small GTPase enriched in cilia. Deletion of Arl13b in mouse kidney results in renal cysts and an associated absence of primary cilia. Similarly, ablation of cilia leads to kidney cysts. To investigate whether ARL13B functions from within cilia to direct kidney development, we examined kidneys of mice expressing an engineered cilia-excluded ARL13B variant, ARL13BV358A. These mice retained renal cilia and developed cystic kidneys. Because ARL13B functions as a guanine nucleotide exchange factor (GEF) for ARL3, we examined kidneys of mice expressing an ARL13B variant that lacks ARL3 GEF activity, ARL13BR79Q. We found normal kidney development with no evidence of cysts in these mice. Taken together, our results show that ARL13B functions within cilia to inhibit renal cystogenesis during mouse development, and that this function does not depend on its role as a GEF for ARL3.

4.
Ann Breast Cancer Ther ; 4(1): 48-57, 2020 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-32542231

RESUMEN

Chemo-resistant breast cancer is a major barrier to curative treatment for a significant number of women with breast cancer. Neoadjuvant chemotherapy (NACT) is standard first- line treatment for most women diagnosed with high-risk TNBC, HER2+, and locally advanced ER+ breast cancer. Current clinical prognostic tools evaluate four clinicopathological factors: Tumor size, LN status, pathological stage, and tumor molecular subtype. However, many similarly treated patients with identical residual cancer burden (RCB) following NACT experience distinctly different tumor relapse rates, clinical outcomes and survival. This problem is particularly apparent for incomplete responders with a high-risk RCB classification following NACT. Therefore, there is a pressing need to identify new prognostic and predictive biomarkers, and develop novel curative therapies to augment current standard of care (SOC) treatment regimens to save more lives. Here, we will discuss these unmet needs and clinical challenges that stand in the way of precision medicine and personalized cancer therapy.

5.
Cancers (Basel) ; 10(5)2018 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-29757973

RESUMEN

Oncogenic K-RAS mutations are found in virtually all pancreatic cancers, making K-RAS one of the most targeted oncoproteins for drug development in cancer therapies. Despite intense research efforts over the past three decades, oncogenic K-RAS has remained largely "undruggable". Rather than targeting an upstream component of the RAS signaling pathway (i.e., EGFR/HER2) and/or the midstream effector kinases (i.e., RAF/MEK/ERK/PI3K/mTOR), we propose an alternative strategy to control oncogenic K-RAS signal by targeting its most downstream signaling module, Seven-In-Absentia Homolog (SIAH). SIAH E3 ligase controls the signal output of oncogenic K-RAS hyperactivation that drives unchecked cell proliferation, uncontrolled tumor growth, and rapid cancer cell dissemination in human pancreatic cancer. Therefore, SIAH is an ideal therapeutic target as it is an extraordinarily conserved downstream signaling gatekeeper indispensable for proper RAS signaling. Guided by molecular insights and core principles obtained from developmental and evolutionary biology, we propose an anti-SIAH-centered anti-K-RAS strategy as a logical and alternative anticancer strategy to dampen uncontrolled K-RAS hyperactivation and halt tumor growth and metastasis in pancreatic cancer. The clinical utility of developing SIAH as both a tumor-specific and therapy-responsive biomarker, as well as a viable anti-K-RAS drug target, is logically simple and conceptually innovative. SIAH clearly constitutes a major tumor vulnerability and K-RAS signaling bottleneck in pancreatic ductal adenocarcinoma (PDAC). Given the high degree of evolutionary conservation in the K-RAS/SIAH signaling pathway, an anti-SIAH-based anti-PDAC therapy will synergize with covalent K-RAS inhibitors and direct K-RAS targeted initiatives to control and eradicate pancreatic cancer in the future.

6.
BMC Evol Biol ; 17(1): 182, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28784114

RESUMEN

BACKGROUND: The RAS signaling pathway is a pivotal developmental pathway that controls many fundamental biological processes including cell proliferation, differentiation, movement and apoptosis. Drosophila Seven-IN-Absentia (SINA) is a ubiquitin E3 ligase that is the most downstream signaling "gatekeeper" whose biological activity is essential for proper RAS signal transduction. Vertebrate SINA homologs (SIAHs) share a high degree of amino acid identity with that of Drosophila SINA. SINA/SIAH is the most conserved signaling component in the canonical EGFR/RAS/RAF/MAPK signal transduction pathway. RESULTS: Vertebrate SIAH1, 2, and 3 are the three orthologs to invertebrate SINA protein. SINA and SIAH1 orthologs are found in all major taxa of metazoans. These proteins have four conserved functional domains, known as RING (Really Interesting New Gene), SZF (SIAH-type zinc finger), SBS (substrate binding site) and DIMER (Dimerization). In addition to the siah1 gene, most vertebrates encode two additional siah genes (siah2 and siah3) in their genomes. Vertebrate SIAH2 has a highly divergent and extended N-terminal sequence, while its RING, SZF, SBS and DIMER domains maintain high amino acid identity/similarity to that of SIAH1. But unlike vertebrate SIAH1 and SIAH2, SIAH3 lacks a functional RING domain, suggesting that SIAH3 may be an inactive E3 ligase. The SIAH3 subtree exhibits a high degree of amino acid divergence when compared to the SIAH1 and SIAH2 subtrees. We find that SIAH1 and SIAH2 are expressed in all human epithelial cell lines examined thus far, while SIAH3 is only expressed in a limited subset of cancer cell lines. CONCLUSION: Through phylogenetic analyses of metazoan SINA and SIAH E3 ligases, we identified many invariant and divergent amino acid residues, as well as the evolutionarily conserved functional motifs in this medically relevant gene family. Our phylomedicinal study of this unique metazoan SINA/SIAH protein family has provided invaluable evolution-based support towards future effort to design logical, potent, and durable anti-SIAH-based anticancer strategies against oncogenic K-RAS-driven metastatic human cancers. Thus, this method of evolutionary study should be of interest in cancer biology.


Asunto(s)
Proteínas Nucleares/clasificación , Filogenia , Ubiquitina-Proteína Ligasas/clasificación , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Secuencia de Consenso , Evolución Molecular , Regulación Neoplásica de la Expresión Génica , Humanos , Invertebrados/enzimología , Familia de Multigenes , Neoplasias/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Vertebrados/metabolismo
7.
EBioMedicine ; 11: 183-198, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27569656

RESUMEN

BACKGROUND: Metastatic breast cancer exhibits diverse and rapidly evolving intra- and inter-tumor heterogeneity. Patients with similar clinical presentations often display distinct tumor responses to standard of care (SOC) therapies. Genome landscape studies indicate that EGFR/HER2/RAS "pathway" activation is highly prevalent in malignant breast cancers. The identification of therapy-responsive and prognostic biomarkers is paramount important to stratify patients and guide therapies in clinical oncology and personalized medicine. METHODS: In this study, we analyzed matched pairs of tumor specimens collected from 182 patients who received neoadjuvant systemic therapies (NST). Statistical analyses were conducted to determine whether EGFR/HER2/RAS pathway biomarkers and clinicopathological predictors, alone and in combination, are prognostic in breast cancer. FINDINGS: SIAH and EGFR outperform ER, PR, HER2 and Ki67 as two logical, sensitive and prognostic biomarkers in metastatic breast cancer. We found that increased SIAH and EGFR expression correlated with advanced pathological stage and aggressive molecular subtypes. Both SIAH expression post-NST and NST-induced changes in EGFR expression in invasive mammary tumors are associated with tumor regression and increased survival, whereas ER, PR, and HER2 were not. These results suggest that SIAH and EGFR are two prognostic biomarkers in breast cancer with lymph node metastases. INTERPRETATION: The discovery of incorporating tumor heterogeneity-independent and growth-sensitive RAS pathway biomarkers, SIAH and EGFR, whose altered expression can be used to estimate therapeutic efficacy, detect emergence of resistant clones, forecast tumor regression, differentiate among partial responders, and predict patient survival in the neoadjuvant setting, has a clear clinical implication in personalizing breast cancer therapy. FUNDING: This work was supported by the Dorothy G. Hoefer Foundation for Breast Cancer Research (A.H. Tang); Center for Innovative Technology (CIT)-Commonwealth Research Commercialization Fund (CRCF) (MF14S-009-LS to A.H. Tang), and National Cancer Institute (CA140550 to A.H. Tang).


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Receptores ErbB/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas ras/metabolismo , Biomarcadores de Tumor , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Progresión de la Enfermedad , Receptores ErbB/genética , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Modelos Biológicos , Terapia Neoadyuvante , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Proteínas Nucleares/genética , Pronóstico , Modelos de Riesgos Proporcionales , Resultado del Tratamiento , Ubiquitina-Proteína Ligasas/genética , Proteínas ras/genética
8.
Proc Natl Acad Sci U S A ; 113(6): 1558-63, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26831109

RESUMEN

Mutations in genes encoding myosin, the molecular motor that powers cardiac muscle contraction, and its accessory protein, cardiac myosin binding protein C (cMyBP-C), are the two most common causes of hypertrophic cardiomyopathy (HCM). Recent studies established that the N-terminal domains (NTDs) of cMyBP-C (e.g., C0, C1, M, and C2) can bind to and activate or inhibit the thin filament (TF). However, the molecular mechanism(s) by which NTDs modulate interaction of myosin with the TF remains unknown and the contribution of each individual NTD to TF activation/inhibition is unclear. Here we used an integrated structure-function approach using cryoelectron microscopy, biochemical kinetics, and force measurements to reveal how the first two Ig-like domains of cMyPB-C (C0 and C1) interact with the TF. Results demonstrate that despite being structural homologs, C0 and C1 exhibit different patterns of binding on the surface of F-actin. Importantly, C1 but not C0 binds in a position to activate the TF by shifting tropomyosin (Tm) to the "open" structural state. We further show that C1 directly interacts with Tm and traps Tm in the open position on the surface of F-actin. Both C0 and C1 compete with myosin subfragment 1 for binding to F-actin and effectively inhibit actomyosin interactions when present at high ratios of NTDs to F-actin. Finally, we show that in contracting sarcomeres, the activating effect of C1 is apparent only once low levels of Ca(2+) have been achieved. We suggest that Ca(2+) modulates the interaction of cMyBP-C with the TF in the sarcomere.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Actinas/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Humanos , Hidrólisis , Modelos Moleculares , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Contracción Muscular/efectos de los fármacos , Subfragmentos de Miosina/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Conejos , Sarcómeros/efectos de los fármacos , Sarcómeros/metabolismo , Relación Estructura-Actividad , Sus scrofa
9.
Clin Biochem ; 47(15): 89-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24976626

RESUMEN

OBJECTIVES: Exosomes are 50-90nm extracellular membrane particles that may mediate trans-cellular communication between cells and tissues. We have reported that human urinary exosomes contain miRNA that are biomarkers for salt sensitivity and inverse salt sensitivity of blood pressure. This study examines exosomal transfer between cultured human renal proximal tubule cells (RPTCs) and from RPTCs to human distal tubule and collecting duct cells. DESIGN AND METHODS: For RPTC-to-RPTC exosomal transfer, we utilized 5 RPTC lines producing exosomes that were fluorescently labeled with exosomal-specific markers CD63-EGFP or CD9-RFP. Transfer between RPTCs was demonstrated by co-culturing CD63-EGFP and CD9-RFP stable clones and performing live confocal microscopy. For RPTC-to-distal segment exosomal transfer, we utilized 5 distal tubule and 3 collecting duct immortalized cell lines. RESULTS: Time-lapse videos revealed unique proximal tubule cellular uptake patterns for exosomes and eventual accumulation into the multivesicular body. Using culture supernatant containing exosomes from 3 CD9-RFP and 2 CD63-EGFP RPTC cell lines, all 5 distal tubule cell lines and all 3 collecting duct cell lines showed exosomal uptake as measured by microplate fluorometry. Furthermore, we found that RPTCs stimulated with fenoldopam (dopamine receptor agonist) had increased production of exosomes, which upon transfer to distal tubule and collecting duct cells, reduced the basal reactive oxygen species (ROS) production rates in those recipient cells. CONCLUSION: Due to the complex diversity of exosomal contents, this proximal-to-distal vesicular inter-nephron transfer may represent a previously unrecognized trans-renal communication system.


Asunto(s)
Exosomas/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Distales/metabolismo , Túbulos Renales Proximales/metabolismo , Comunicación Celular , Línea Celular , Exosomas/genética , Humanos , Riñón/citología , Riñón/metabolismo , Túbulos Renales Colectores/citología , Túbulos Renales Distales/citología , Túbulos Renales Proximales/citología , MicroARNs/genética , Nefronas/metabolismo , Tetraspanina 29/genética , Tetraspanina 30/genética
10.
Kidney Int ; 86(1): 118-26, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24552847

RESUMEN

Determining the individual roles of the two dopamine D1-like receptors (D1R and D5R) on sodium transport in the human renal proximal tubule has been complicated by their structural and functional similarity. Here we used a novel D5R-selective antagonist (LE-PM436) and D1R- or D5R-specific gene silencing to determine second messenger coupling pathways and heterologous receptor interaction between the two receptors. D1R and D5R colocalize in renal proximal tubule cells and physically interact, as determined by co-immunoprecipitation and fluorescent resonance energy transfer microscopy. Stimulation of renal proximal tubule cells with fenoldopam (D1R/D5R agonist) led to both adenylyl cyclase and phospholipase C (PLC) activation using real-time fluorescent resonance energy transfer biosensors ICUE3 and CYPHR, respectively. Fenoldopam increased cAMP accumulation and PLC activity and inhibited both NHE3 and NaKATPase activities. LE-PM436 and D5R siRNA blocked the fenoldopam-stimulated PLC pathway but not cAMP accumulation, whereas D1R siRNA blocked both fenoldopam-stimulated cAMP accumulation and PLC signaling. Either D1R or D5R siRNA, or LE-PM436 blocked the fenoldopam-dependent inhibition of sodium transport. Further studies using the cAMP-selective D1R/D5R agonist SKF83822 and PLC-selective D1R/D5R agonist SKF83959 confirmed the cooperative influence of the two pathways on sodium transport. Thus, D1R and D5R interact in the inhibition of NHE3 and NaKATPase activity, the D1R primarily by cAMP, whereas the D1R/D5R heteromer modulates the D1R effect through a PLC pathway.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D5/metabolismo , Sodio/metabolismo , Benzazepinas/farmacología , Transporte Biológico Activo/efectos de los fármacos , Células Cultivadas , Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Fenoldopam/farmacología , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Modelos Biológicos , ARN Interferente Pequeño/genética , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D5/agonistas , Receptores de Dopamina D5/antagonistas & inhibidores , Sistemas de Mensajero Secundario
11.
Clin Chim Acta ; 421: 236-42, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23454474

RESUMEN

BACKGROUND: Salt sensitivity (SS) of blood pressure (BP) affects 25% of adults, shares comorbidity with hypertension, and has no convenient diagnostic test. We tested the hypothesis that urine-derived exfoliated renal proximal tubule cells (RPTCs) could diagnose the degree of an individual's SS of BP. METHODS: Subjects were selected who had their SS of BP determined 5 y prior to this study (salt-sensitive: ≥7 mm Hg increase in mean arterial pressure (MAP) following transition from a random weekly diet of low (10 mmol/day) to high (300 mmol/day) sodium (Na(+)) intake, N=4; inverse salt-sensitive (ISS): ≥7 mm Hg increase in MAP transitioning from a high to low Na(+) diet, N=3, and salt-resistant (SR): <7 mm Hg change in MAP transitioned on either diet, N=5). RPTC responses to 2 independent Na(+) transport pathways were measured. RESULTS: There was a negative correlation between the degree of SS and dopamine-1 receptor (D1R) plasma membrane recruitment (y=-0.0107x+0.68 relative fluorescent units (RFU), R(2)=0.88, N=12, P<0.0001) and angiotensin II-stimulated intracellular Ca(++) (y=-0.0016x+0.0336, R(2)=0.7112, P<0.001, N=10) concentration over baseline. CONCLUSIONS: Isolating RPTCs from urine provides a personalized cell-based diagnostic test of SS index that offers advantages over a 2-week controlled diet with respect to cost and patient compliance. Furthermore, the linear relationship between the change in MAP and response to 2 Na(+) regulatory pathways suggests that an individual's RPTC response to intracellular Na(+) is personalized and predictive.


Asunto(s)
Presión Arterial/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Sodio en la Dieta/farmacología , Angiotensina II/genética , Angiotensina II/metabolismo , Biomarcadores/metabolismo , Calcio/metabolismo , Separación Celular , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Sodio en la Dieta/metabolismo
12.
Hypertension ; 61(5): 1021-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23509080

RESUMEN

The G protein-coupled receptor kinase 4 (GRK4) negatively regulates the dopaminergic system by desensitizing the dopamine-1-receptor. The expressional control of GRK4 has not been reported, but here we show that the transcription factor c-Myc binds to the promoter of GRK4 and positively regulates GRK4 protein expression in human renal proximal tubule cells (RPTCs). Addition of phorbol esters to RPTCs not only increased c-Myc binding to the GRK4 promoter but also increased both phospho-c-Myc and GRK4 expression. The phorbol ester-mediated increase in GRK4 expression was completely blocked by the c-Myc inhibitor, 10074-G5, indicating that GRK4 is downstream of phospho-c-Myc. The autocrine production of angiotensin II (Ang II) in RPTCs increased the phosphorylation and activation of c-Myc and subsequently GRK4 expression. 3-Amino-4-thio-butyl sulfonate, an inhibitor of aminopeptidase A, increased RPTC secretion of Ang II. 3-Amino-4-thio-butyl sulfonate or Ang II increased the expression of both phospho-c-Myc and GRK4, which was blocked by 10074-G5. Blockade of the Ang II type 1 receptor with losartan decreased phospho-c-Myc and GRK4 expression. Both inhibition of c-Myc activity and blockade of Ang II type 1 receptor restored the coupling of dopamine-1-receptor to adenylyl cyclase stimulation in uncoupled RPTCs, whereas phorbol esters or Ang II caused the uncoupling of normally coupled RPTCs. We suggest that the Ang II type 1 receptor impairs dopamine-1-receptor function via c-Myc activation of GRK4. This novel pathway may be involved in the increase in blood pressure in hypertension that is mediated by increased activity of the renin-angiotensin system and decreased activity of the renal dopaminergic system.


Asunto(s)
Quinasa 4 del Receptor Acoplado a Proteína-G/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética/fisiología , Angiotensina II/metabolismo , Línea Celular , Células Cultivadas , Quinasa 4 del Receptor Acoplado a Proteína-G/genética , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Losartán/farmacología , Oxadiazoles/farmacología , Ésteres del Forbol/farmacología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Dopaminérgicos/metabolismo , Transcripción Genética/efectos de los fármacos
13.
Methods Mol Biol ; 945: 329-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23097116

RESUMEN

The kidney is a highly heterogeneous organ that is responsible for fluid and electrolyte balance. Much interest is focused on determining the function of specific renal epithelial cells in humans, which can only be accomplished through the isolation and growth of nephron segment-specific epithelial cells. However, human renal epithelial cells are notoriously difficult to maintain in culture. This chapter describes the isolation, growth, immortalization, and characterization of the human renal proximal tubule cell. In addition, we describe new paradigms in 3D cell culture which allow the cells to maintain more in vivo-like morphology and function.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Células Epiteliales/citología , Túbulos Renales Proximales/citología , Adenoviridae/genética , Antígenos Transformadores de Poliomavirus/genética , Proliferación Celular , Criopreservación , Células Epiteliales/metabolismo , Células HEK293 , Humanos , Lentivirus/genética , Telomerasa/genética , Transducción Genética
14.
Am J Physiol Renal Physiol ; 300(4): F914-20, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21289050

RESUMEN

Renal dopamine receptor function and ion transport inhibition are impaired in essential hypertension. We recently reported that caveolin-1 (CAV1) and lipid rafts are necessary for normal D(1)-like receptor-dependent internalization of Na-K-ATPase in human proximal tubule cells. We now hypothesize that CAV1 is necessary for the regulation of urine sodium (Na(+)) excretion (U(Na)V) and mean arterial blood pressure (MAP) in vivo. Acute renal interstitial (RI) infusion into Sprague-Dawley rats of 1 µg·kg⁻¹·min⁻¹ fenoldopam (FEN; D(1)-like receptor agonist) caused a 0.46 ± 0.15-µmol/min increase in U(Na)V (over baseline of 0.29 ± 0.04 µmol/min; P < 0.01). This increase was seen in Na(+)-loaded rats, but not in those under a normal-sodium load. Coinfusion with ß-methyl cyclodextrin (ßMCD; lipid raft disrupter, 200 µg·kg⁻¹·min⁻¹) completely blocked this FEN-induced natriuresis (P < 0.001). Long-term (3 day) lipid raft disruption via continuous RI infusion of 80 µg·kg⁻¹·min⁻¹ ßMCD decreased renal cortical CAV1 expression (47.3 ± 6.4%; P < 0.01) and increased MAP (32.4 ± 6.6 mmHg; P < 0.001) compared with vehicle-infused animals. To determine whether the MAP rise was due to a CAV1-dependent lipid raft-mediated disruption, Na(+)-loaded rats were given a bolus RI infusion of CAV1 siRNA. Two days postinfusion, cortical CAV1 expression was decreased by 73.6 ± 8.2% (P < 0.001) and the animals showed an increase in MAP by 17.4 ± 2.9 mmHg (P < 0.01) compared with animals receiving scrambled control siRNA. In summary, acute kidney-specific lipid raft disruption decreases CAV1 expression and blocks D(1)-like receptor-induced natriuresis. Furthermore, chronic disruption of lipid rafts or CAV1 protein expression in the kidney induces hypertension.


Asunto(s)
Caveolina 1/metabolismo , Hipertensión/metabolismo , Riñón/metabolismo , Natriuresis/fisiología , Sodio en la Dieta/metabolismo , Análisis de Varianza , Animales , Caveolina 1/genética , Femenino , Técnica del Anticuerpo Fluorescente , Hipertensión/fisiopatología , Inmunoprecipitación , Riñón/fisiopatología , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Sodio en la Dieta/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA