Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Clin Invest ; 54(7): e14177, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38381498

RESUMEN

BACKGROUND: The role of insulin resistance in hepatic fibrosis in Metabolic dysfunction-Associated SteatoHepatitis (MASH) remains unclear. Carcinoembryonic Antigen-related Cell Adhesion Molecule1 protein (CEACAM1) promotes insulin clearance to maintain insulin sensitivity and repress de novo lipogenesis, as bolstered by the development of insulin resistance and steatohepatitis in AlbuminCre + Cc1fl/fl mice with liver-specific mouse gene encoding CEACAM1 protein (Ceacam1) deletion. We herein investigated whether these mice also developed hepatic fibrosis and whether hepatic CEACAM1 is reduced in patients with MASH at different fibrosis stages. METHODS: AlbuminCre + Cc1fl/fl mice were fed a regular or a high-fat diet before their insulin metabolism and action were assessed during IPGTT, and their livers excised for histochemical, immunohistochemical and Western blot analysis. Sirius red staining was used to assess fibrosis, and media transfer was employed to examine whether mutant hepatocytes activated hepatic stellate cells (HSCs). Hepatic CEACAM1 protein levels in patients with varying disease stages were assessed by ELISA. RESULTS: Hepatocytic deletion of Ceacam1 caused hyperinsulinemia-driven insulin resistance emanating from reduced hepatic insulin clearance. AlbuminCre + Cc1fl/fl livers showed inflammation, fibrosis and hepatic injury, with more advanced bridging and chicken-wire hepatic fibrosis under high-fat conditions. Media transferred from hepatocytes isolated from mutant mice activated control HSCs, likely owing to their elevated endothelin1 content. Interestingly, hepatic CEACAM1 levels were lower in the livers of patients with MASH and declined gradually with advanced fibrosis stage. CONCLUSIONS: Hepatic CEACAM1 levels declined with progression of MASH in humans. The phenotype of AlbuminCre + Cc1fl/fl mice assigned a key role to CEACAM1 loss from hepatocytes in hepatic fibrosis independently of other liver cells.


Asunto(s)
Hepatocitos , Resistencia a la Insulina , Cirrosis Hepática , Animales , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Ratones , Humanos , Resistencia a la Insulina/fisiología , Dieta Alta en Grasa , Antígeno Carcinoembrionario/metabolismo , Masculino , Células Estrelladas Hepáticas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/genética , Hiperinsulinismo/metabolismo , Hígado Graso/metabolismo , Antígenos CD/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
2.
Nat Commun ; 14(1): 4250, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460527

RESUMEN

Defects in insulin processing and granule maturation are linked to pancreatic beta-cell failure during type 2 diabetes (T2D). Phosphatidylinositol transfer protein alpha (PITPNA) stimulates activity of phosphatidylinositol (PtdIns) 4-OH kinase to produce sufficient PtdIns-4-phosphate (PtdIns-4-P) in the trans-Golgi network to promote insulin granule maturation. PITPNA in beta-cells of T2D human subjects is markedly reduced suggesting its depletion accompanies beta-cell dysfunction. Conditional deletion of Pitpna in the beta-cells of Ins-Cre, Pitpnaflox/flox mice leads to hyperglycemia resulting from decreasing glucose-stimulated insulin secretion (GSIS) and reducing pancreatic beta-cell mass. Furthermore, PITPNA silencing in human islets confirms its role in PtdIns-4-P synthesis and leads to impaired insulin granule maturation and docking, GSIS, and proinsulin processing with evidence of ER stress. Restoration of PITPNA in islets of T2D human subjects reverses these beta-cell defects and identify PITPNA as a critical target linked to beta-cell failure in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proinsulina/metabolismo
3.
Biomedicines ; 11(1)2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36672619

RESUMEN

The genetic and molecular basis of developing high blood pressure and renal disease are not well known. Resp18mutant Dahl salt-sensitive (SS-Resp18mutant) rats fed a 2% NaCl diet for six weeks have high blood pressure, increased renal fibrosis, and decreased mean survival time. Impairment of the dopaminergic system also leads to hypertension that involves renal and non-renal mechanisms. Deletion of any of the five dopamine receptors may lead to salt-sensitive hypertension. Therefore, we investigated the interaction between Resp18 and renal dopamine in SS-Resp18mutant and Dahl salt-sensitive (SS) rats. We found that SS-Resp18mutant rats had vascular dysfunction, as evidenced by a decrease in vasorelaxation in response to sodium nitroprusside. The pressure-natriuresis curve in SS-Resp18mutant rats was shifted down and to the right of SS rats. SS-Resp18mutant rats had decreased glomerular filtration rate and dopamine receptor subtypes, D1R and D5R. Renal dopamine levels were decreased, but urinary dopamine levels were increased, which may be the consequence of increased renal dopamine production, followed by secretion into the tubular lumen. The increased renal dopamine production in SS-Resp18mutant rats in vivo was substantiated by the increased dopamine production in renal proximal tubule cells treated with L-DOPA. Overall, our study provides evidence that targeted disruption of the Resp18 locus in the SS rat dysregulates the renal dopaminergic system.

4.
Biomedicines ; 10(8)2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-36009446

RESUMEN

Insulin stores lipid in adipocytes and prevents lipolysis and the release of non-esterified fatty acids (NEFA). Excessive release of NEFA during sustained energy supply and increase in abdominal adiposity trigger systemic insulin resistance, including in the liver, a major site of insulin clearance. This causes a reduction in insulin clearance as a compensatory mechanism to insulin resistance in obesity. On the other hand, reduced insulin clearance in the liver can cause chronic hyperinsulinemia, followed by downregulation of insulin receptor and insulin resistance. Delineating the cause-effect relationship between reduced insulin clearance and insulin resistance has been complicated by the fact that insulin action and clearance are mechanistically linked to insulin binding to its receptors. This review discusses how NEFA mobilization contributes to the reciprocal relationship between insulin resistance and reduced hepatic insulin clearance, and how this may be implicated in the pathogenesis of non-alcoholic fatty liver disease.

5.
Neurooncol Adv ; 4(1): vdac095, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35875691

RESUMEN

Background: The prognosis of glioblastoma (GBM) remains dismal because therapeutic approaches have limited effectiveness. A new targeted treatment using MEK inhibitors, including trametinib, has been proposed to improve GBM therapy. Trametinib had a promising preclinical effect against several cancers, but its adaptive treatment resistance precluded its clinical translation in GBM. Previously, we have demonstrated that protein arginine methyltransferase 5 (PRMT5) is upregulated in GBM and its inhibition promotes apoptosis and senescence in differentiated and stem-like tumor cells, respectively. We tested whether inhibition of PRMT5 can enhance the efficacy of trametinib against GBM. Methods: Patient-derived primary GBM neurospheres (GBMNS) with transient PRMT5 knockdown were treated with trametinib and cell viability, proliferation, cell cycle progression, ELISA, and western blot were analyzed. In vivo, NSG mice were intracranially implanted with PRMT5-intact and -depleted GBMNS, treated with trametinib by daily oral gavage, and observed for tumor progression and mice survival rate. Results: PRMT5 depletion enhanced trametinib-induced cytotoxicity in GBMNS. PRMT5 knockdown significantly decreased trametinib-induced AKT and ERBB3 escape pathways. However, ERBB3 inhibition alone failed to block trametinib-induced AKT activity suggesting that the enhanced antitumor effect imparted by PRMT5 knockdown in trametinib-treated GBMNS resulted from AKT inhibition and not ERBB3 inhibition. In orthotopic murine xenograft models, PRMT5-depletion extended the survival of tumor-bearing mice, and combination with trametinib further increased survival. Conclusion: Combined PRMT5/MEK inhibition synergistically inhibited GBM in animal models and is a promising strategy for GBM therapy.

6.
Biochem Biophys Res Commun ; 572: 35-40, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34340197

RESUMEN

Expression of Regulated endocrine specific protein 18 (Resp18) is localized in numerous tissues and cell types; however, its exact cellular function is unknown. We previously showed that targeted disruption of the Resp18 locus in the Dahl SS (SS) rat (Resp18mutant) results in higher blood pressure (BP), increased renal fibrosis, increased urinary protein excretion, and decreased mean survival time following a chronic (6 weeks) 2% high salt (HS) diet compared with the SS rat. Based on this prominent renal injury phenotype, we hypothesized that targeted disruption of Resp18 in the SS rat promotes an early onset hypertensive-signaling event through altered signatures of the renal transcriptome in response to HS. To test this hypothesis, both SS and Resp18mutant rats were exposed to a 7-day 2% HS diet and BP was recorded by radiotelemetry. After a 7-day exposure to the HS diet, systolic BP was significantly increased in the Resp18mutant rat compared with the SS rat throughout the circadian cycle. Therefore, we sought to investigate the renal transcriptomic response to HS in the Resp18mutant rat. Using RNA sequencing, Resp18mutant rats showed a differential expression of 25 renal genes, including upregulation of Ren. Upregulation of renal Ren and other differentially expressed genes were confirmed via qRT-PCR. Moreover, circulating renin activity was significantly higher in the Resp18mutant rat compared with the WT SS rat after 7 days on HS. Collectively, these observations demonstrate that disruption of the Resp18 gene in the SS rat is associated with an altered renal transcriptomics signature as an early response to salt load.


Asunto(s)
Riñón/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Perfilación de la Expresión Génica , Masculino , Mutación , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Endogámicas Dahl
7.
J Am Heart Assoc ; 9(7): e014072, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32200719

RESUMEN

Background Renal artery stenosis is a common cause of renal ischemia, contributing to the development of chronic kidney disease. To investigate the role of local CD40 expression in renal artery stenosis, Goldblatt 2-kidney 1-clip surgery was performed on hypertensive Dahl salt-sensitive rats (S rats) and genetically modified S rats in which CD40 function is abolished (Cd40mutant). Methods and Results Four weeks following the 2-kidney 1-clip procedure, Cd40mutant rats demonstrated significantly reduced blood pressure and renal fibrosis in the ischemic kidneys compared with S rat controls. Similarly, disruption of Cd40 resulted in reduced 24-hour urinary protein excretion in Cd40mutant rats versus S rat controls (46.2±1.9 versus 118.4±5.3 mg/24 h; P<0.01), as well as protection from oxidative stress, as indicated by increased paraoxonase activity in Cd40mutant rats versus S rat controls (P<0.01). Ischemic kidneys from Cd40mutant rats demonstrated a significant decrease in gene expression of the profibrotic mediator, plasminogen activator inhibitor-1 (P<0.05), and the proinflammatory mediators, C-C motif chemokine ligand 19 (P<0.01), C-X-C Motif Chemokine Ligand 9 (P<0.01), and interleukin-6 receptor (P<0.001), compared with S rat ischemic kidneys, as assessed by quantitative PCR assay. Reciprocal renal transplantation documented that CD40 exclusively expressed in the kidney contributes to ischemia-induced renal fibrosis. Furthermore, human CD40-knockout proximal tubule epithelial cells suggested that suppression of CD40 signaling significantly inhibited expression of proinflammatory and -fibrotic genes. Conclusions Taken together, our data suggest that activation of CD40 induces a significant proinflammatory and -fibrotic response and represents an attractive therapeutic target for treatment of ischemic renal disease.


Asunto(s)
Antígenos CD40/metabolismo , Isquemia/metabolismo , Riñón/irrigación sanguínea , Riñón/metabolismo , Mutación , Obstrucción de la Arteria Renal/metabolismo , Animales , Presión Sanguínea , Antígenos CD40/genética , Línea Celular , Modelos Animales de Enfermedad , Fibrosis , Tasa de Filtración Glomerular , Humanos , Mediadores de Inflamación/metabolismo , Isquemia/genética , Isquemia/patología , Isquemia/fisiopatología , Riñón/patología , Riñón/fisiopatología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Estrés Oxidativo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas Endogámicas Dahl , Obstrucción de la Arteria Renal/genética , Obstrucción de la Arteria Renal/patología , Obstrucción de la Arteria Renal/fisiopatología , Transducción de Señal
8.
Physiol Genomics ; 52(1): 1-14, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31762410

RESUMEN

Here we postulate that the heritability of complex disease traits previously ascribed solely to the inheritance of the nuclear and mitochondrial genomes is broadened to encompass a third component of the holobiome, the microbiome. To test this, we expanded on the selectively bred low capacity runner/high capacity runner (LCR/HCR) rat exercise model system into four distinct rat holobiont model frameworks including matched and mismatched host nuclear and mitochondrial genomes. Vertical selection of varying nuclear and mitochondrial genomes resulted in differential acquisition of the microbiome within each of these holobiont models. Polygenic disease risk of these novel models were assessed and subsequently correlated with patterns of acquisition and contributions of their microbiomes in controlled laboratory settings. Nuclear-mitochondrial-microbiotal interactions were not for exercise as a reporter of health, but significantly noted for increased adiposity, increased blood pressure, compromised cardiac function, and loss of long-term memory as reporters of disease susceptibility. These findings provide evidence for coselection of the microbiome with nuclear and mitochondrial genomes as an important feature impacting the heritability of complex diseases.


Asunto(s)
Núcleo Celular/genética , Microbioma Gastrointestinal/genética , Predisposición Genética a la Enfermedad , Genoma Mitocondrial , Adiposidad/genética , Animales , Conducta Animal , Presión Sanguínea/genética , Peso Corporal/genética , Enfermedades Cardiovasculares/genética , Cognición , ADN Mitocondrial/genética , Condicionamiento Físico Animal , Ratas , Factores de Riesgo , Selección Genética , Remodelación Ventricular/genética
9.
Endocrinology ; 160(9): 2093-2100, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31294787

RESUMEN

Regulated endocrine-specific protein-18 (RESP18), a novel 18-kDa protein, was first identified in neuroendocrine tissue. Subsequent studies showed that Resp18 is expressed in the adrenal medulla, brain, pancreas, pituitary, retina, stomach, superior cervical ganglion, testis, and thyroid and also circulates in the plasma. Resp18 has partial homology with the islet cell antigen 512, also known as protein tyrosine phosphatase, receptor type N (PTPRN), but does not have phosphatase activity. Resp18 might serve as an intracellular signal; however, its function is unclear. It is regulated by dopamine, glucocorticoids, and insulin. We recently reported that the targeted disruption of the Resp18 locus in Dahl salt-sensitive rats increased their blood pressure and caused renal injury. The aim of the present review was to provide a comprehensive summary of the reported data currently available, especially the expression and proposed organ-specific function of Resp18.


Asunto(s)
Proteínas del Tejido Nervioso/fisiología , Animales , Enfermedades Cardiovasculares/prevención & control , Dopamina/fisiología , Gastrinas/fisiología , Glucocorticoides/fisiología , Humanos , Sistema Hipotálamo-Hipofisario/fisiología , Insulina/fisiología , Proteínas del Tejido Nervioso/química , Enfermedad de Parkinson/etiología
10.
Biochem Pharmacol ; 168: 71-81, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31228465

RESUMEN

Iron is essential for many biological functions, including being a cofactor for enzymes involved in cell proliferation. In line, it has been shown that cancer cells can perturb their iron metabolism towards retaining an abundant iron supply for growth and survival. Accordingly, it has been suggested that iron deprivation through the use of iron chelators could attenuate cancer progression. While they have exhibited anti-tumor properties in vitro, the current therapeutic iron chelators are inadequate due to their low efficacy. Therefore, we investigated whether the bacterial catecholate-type siderophore, enterobactin (Ent), could be used as a potent anti-cancer agent given its strong iron chelation property. We demonstrated that iron-free Ent can exert cytotoxic effects specifically towards monocyte-related tumor cell lines (RAW264.7 and J774A.1), but not primary cells, i.e. bone marrow-derived macrophages (BMDMs), through two mechanisms. First, we observed that RAW264.7 and J774A.1 cells preserve a bountiful intracellular labile iron pool (LIP), whose homeostasis can be disrupted by Ent. This may be due, in part, to the lower levels of lipocalin 2 (Lcn2; an Ent-binding protein) in these cell lines, whereas the higher levels of Lcn2 in BMDMs could prevent Ent from hindering their LIP. Secondly, we observed that Ent could dose-dependently impede reactive oxygen species (ROS) generation in the mitochondria. Such disruption in LIP balance and mitochondrial function may in turn promote cancer cell apoptosis. Collectively, our study highlights Ent as an anti-cancer siderophore, which can be exploited as an unique agent for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Enterobactina/farmacología , Sideróforos/farmacología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Escherichia coli/química , Homeostasis/efectos de los fármacos , Hierro/metabolismo , Lipocalina 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
11.
Biochem Biophys Res Commun ; 513(3): 760-765, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30992131

RESUMEN

Several independent genome-wide association studies (GWAS) have indicated that calcium (Ca2+) voltage-gated channel auxiliary subunit beta 2 (CACNB2) an L-type Ca2+ channel (LTCC) associated protein has strong association with hypertension. However, the molecular mechanism of CACNB2 and its role in the pathophysiology of hypertension is not clear. To address this knowledge gap, we utilized in vitro and in vivo approaches using HEK293 cells and genetically hypertensive, Dahl Salt-Sensitive (SS) rats. We demonstrated that CACNB2 over-expression in HEK293 cells triggers cell proliferation via an up-regulation of the RAS-MAPK pathway compared to non-transfected cells. These effects were likely independent of LTCC activity as treatment with nifedipine, a well-known LTCC blocker, in CACNB2 overexpressing cells failed to inhibit the RAS-MAPK pathway gene expressions or show an effect on apoptosis marker gene expression. Furthermore, the expression level of CACNB2 was up-regulated in the high salt (HS) diet fed SS rat kidneys compared to low salt diet (LS) fed group. Similar to our in vitro observation the RAS-MAPK mRNA levels were increased in HS fed SS rat kidneys, compared to LS fed group. Collectively, our data suggest that CACNB2 is associated with the increase in RAS-MAPK gene expressions and lead us to speculate that in addition to its role in regulating LTCC α1-subunit trafficking, CACNB2 might lead to aberrant RAS activation, which is one of the key cascade associated with hypertension.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Hipertensión/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal , Cloruro de Sodio Dietético/metabolismo , Proteínas ras/metabolismo , Animales , Canales de Calcio Tipo L/genética , Células HEK293 , Humanos , Hipertensión/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Ratas , Ratas Endogámicas Dahl , Regulación hacia Arriba , Proteínas ras/genética
12.
J Enzyme Inhib Med Chem ; 34(1): 863-876, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30915862

RESUMEN

The present study describes the synthesis of a series of 22 chalcone analogs. These compounds were evaluated as potential human MAO-A and MAO-B inhibitors. The compounds showed varied selectivity against the two isoforms. The IC50 values were found to be in the micromolar to submicromolar range. The Ki values of compound 16 were determined to be 0.047 and 0.020 µM for the inhibition of MAO-A and MAO-B, respectively. Dialysis of enzyme-inhibitor mixtures indicated a reversible competitive mode of inhibition. Most of the synthesized chalcone analogs showed a better selectivity toward MAO-B. However, introducing of 2,4,6-trimethoxy substituents on ring B shifted the selectivity toward MAO-A. In addition, we investigated the molecular mechanism of MAO-B inhibition by selected chalcone analogs. Our results revealed that these selected chalcone analogs increased dopamine levels in the rat hepatoma (H4IIE) cells and decreased the relative mRNA expression of the MAO-B enzyme.


Asunto(s)
Chalcona/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Chalcona/síntesis química , Chalcona/química , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de la Monoaminooxidasa/síntesis química , Inhibidores de la Monoaminooxidasa/química , Relación Estructura-Actividad
13.
Steroids ; 141: 63-69, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30481528

RESUMEN

Chicken Ovalbumin Upstream Promoter Transcription Factor II (COUP-TFII) is an orphan member of the nuclear receptor family of transcriptional regulators. Although hormonal activation of COUP-TFII has not yet been identified, rodent genetic models have uncovered vital and diverse roles for COUP-TFII in biological processes. These include control of cardiac function and angiogenesis, reproduction, neuronal development, cell fate and organogenesis. Recently, an emerging body of evidence has demonstrated COUP-TFII involvement in various metabolic systems such as adipogenesis, lipid metabolism, hepatic gluconeogenesis, insulin secretion, and regulation of blood pressure. The potential relevance of these observations to human pathology has been corroborated by the identification of single nucleotide polymorphism in the human COUP-TFII promoter controlling insulin sensitivity. Of particular interest to metabolism is the ability of COUP-TFII to interact with the Glucocorticoid Receptor (GR). This interaction is known to control gluconeogenesis, principally through direct binding of COUP-TFII/GR complexes to the promoters of gluconeogenic enzyme genes. However, it is likely that this interaction is critical to other metabolic processes, since GR, like COUP-TFII, is an essential regulator of adipogenesis, insulin sensitivity, and blood pressure. This review will highlight these unique roles of COUP-TFII in metabolic gene regulation.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Animales , Factor de Transcripción COUP II/genética , Regulación de la Expresión Génica , Humanos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
14.
Int J Mol Sci ; 19(7)2018 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-30037072

RESUMEN

Non-coding RNAs are important regulators of protein-coding genes. The current study characterized an antisense long non-coding RNA, ATP1A1-AS1, which is located on the opposite strand of the Na/K-ATPase α1 gene. Our results show that four splice variants are expressed in human adult kidney cells (HK2 cells) and embryonic kidney cells (HEK293 cells). These variants can be detected in both cytosol and nuclear fractions. We also found that the inhibition of DNA methylation has a differential effect on the expression of ATP1A1-AS1 and its sense gene. To investigate the physiological role of this antisense gene, we overexpressed the ATP1A1-AS1 transcripts, and examined their effect on Na/K-ATPase expression and related signaling function in human kidney cells. The results showed that overexpression of the ATP1A1-AS1-203 transcript in HK2 cells reduced the Na/K-ATPase α1 (ATP1A1) gene expression by approximately 20% (p < 0.05), while reducing the Na/K-ATPase α1 protein synthesis by approximately 22% (p < 0.05). Importantly, overexpression of the antisense RNA transcript attenuated ouabain-induced Src activation in HK2 cells. It also inhibited the cell proliferation and potentiated ouabain-induced cell death. These results demonstrate that the ATP1A1-AS1 gene is a moderate negative regulator of Na/K-ATPase α1, and can modulate Na/K-ATPase-related signaling pathways in human kidney cells.


Asunto(s)
Riñón/metabolismo , ARN sin Sentido/metabolismo , ARN Largo no Codificante/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Western Blotting , Línea Celular , Proliferación Celular/genética , Proliferación Celular/fisiología , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Células HEK293 , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/genética
15.
Physiol Genomics ; 50(5): 369-375, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29570433

RESUMEN

Hypertension is a classic example of a complex polygenic trait, impacted by quantitative trait loci (QTL) containing candidate genes thought to be responsible for blood pressure (BP) control in mammals. One such mapped locus is on rat chromosome 9, wherein the proof for a positional candidate gene, regulated endocrine-specific protein-18 ( Resp18) is currently inadequate. To ascertain the status of Resp18 as a BP QTL, a custom targeted gene disruption model of Resp18 was developed on the Dahl salt-sensitive (SS) background. As a result of this zinc-finger nuclease (ZFN)-mediated disruption, a 7 bp deletion occurred within exon 3 of the Resp18 locus. Targeted disruption of Resp18 gene locus in SS rats decreases its gene expression in both heart and kidney tissues regardless of their dietary salt level. Under a high-salt dietary regimen, both systolic and diastolic BP of Resp18mutant rats were significantly increased compared with SS rats. Resp18mutant rats demonstrated increased renal damage, as evidenced by higher proteinuria and increased renal fibrosis compared with SS rats. Furthermore, under a high-salt diet regimen, the mean survival time of Resp18mutant rats was significantly reduced compared with SS rats. These findings serve as evidence in support of Resp18 as a gene associated with the development of hypertension and renal disease.


Asunto(s)
Hipertensión/genética , Enfermedades Renales/genética , Proteínas del Tejido Nervioso/genética , Cloruro de Sodio Dietético/efectos adversos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Presión Sanguínea/genética , Expresión Génica/efectos de los fármacos , Marcación de Gen/métodos , Hipertensión/etiología , Estimación de Kaplan-Meier , Enfermedades Renales/etiología , Sitios de Carácter Cuantitativo/genética , Ratas Endogámicas Dahl , Ratas Mutantes , Eliminación de Secuencia , Cloruro de Sodio Dietético/administración & dosificación
16.
Sci Rep ; 7: 39867, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28051144

RESUMEN

In previous studies using mice with macrophage-specific loss of TRPC3 we found a significant, selective effect of TRPC3 on the biology of M1, or inflammatory macrophages. Whereas activation of some components of the unfolded protein response and the pro-apoptotic mediators CamkII and Stat1 was impaired in Trpc3-deficient M1 cells, gathering insight about other molecular signatures within macrophages that might be affected by Trpc3 expression requires an alternative approach. In the present study we conducted RNA-seq analysis to interrogate the transcriptome of M1 macrophages derived from mice with macrophage-specific loss of TRPC3 and their littermate controls. We identified 160 significantly differentially expressed genes between the two groups, of which 62 were upregulated and 98 downregulated in control vs. Trpc3-deficient M1 macrophages. Gene ontology analysis revealed enrichment in processes associated to cellular movement and lipid signaling, whereas the enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways included networks for calcium signaling and cell adhesion molecules, among others. This is the first deep transcriptomic analysis of macrophages in the context of Trpc3 deficiency and the data presented constitutes a unique resource to further explore functions of TRPC3 in macrophage biology.


Asunto(s)
Perfilación de la Expresión Génica , Macrófagos/metabolismo , Canales Catiónicos TRPC/genética , Animales , Células de la Médula Ósea/citología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/farmacología , Regulación hacia Abajo , Macrófagos/citología , Ratones , Ratones Noqueados , Netrina-1/farmacología , Canales Catiónicos TRPC/deficiencia , Transcriptoma , Regulación hacia Arriba
17.
Kidney Int ; 91(2): 365-374, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27692815

RESUMEN

High blood pressure is a common cause of chronic kidney disease. Because CD40, a member of the tumor necrosis factor receptor family, has been linked to the progression of kidney disease in ischemic nephropathy, we studied the role of Cd40 in the development of hypertensive renal disease. The Cd40 gene was mutated in the Dahl S genetically hypertensive rat with renal disease by targeted-gene disruption using zinc-finger nuclease technology. These rats were then given low (0.3%) and high (2%) salt diets and compared. The resultant Cd40 mutants had significantly reduced levels of both urinary protein excretion (41.8 ± 3.1 mg/24 h vs. 103.7 ± 4.3 mg/24 h) and plasma creatinine (0.36 ± 0.05 mg/dl vs. 1.15 ± 0.19 mg/dl), with significantly higher creatinine clearance compared with the control S rats (3.04 ± 0.48 ml/min vs. 0.93 ± 0.15 ml/min), indicating renoprotection was conferred by mutation of the Cd40 locus. Furthermore, the Cd40 mutants had a significant attenuation in renal fibrosis, which persisted on the high salt diet. However, there was no difference in systolic blood pressure between the control and Cd40 mutant rats. Thus, these data serve as the first evidence for a direct link between Cd40 and hypertensive nephropathy. Hence, renal fibrosis is one of the underlying mechanisms by which Cd40 plays a crucial role in the development of hypertensive renal disease.


Asunto(s)
Presión Sanguínea/genética , Antígenos CD40/genética , Hipertensión/genética , Enfermedades Renales/prevención & control , Riñón/metabolismo , Mutación , Proteinuria/prevención & control , Animales , Linfocitos B/metabolismo , Antígenos CD40/metabolismo , Movimiento Celular , Creatinina/sangre , Dieta Hiposódica , Modelos Animales de Enfermedad , Fibrosis , Predisposición Genética a la Enfermedad , Hipertensión/metabolismo , Hipertensión/fisiopatología , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Activación de Linfocitos , Fenotipo , Fosforilación , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteinuria/genética , Proteinuria/metabolismo , Proteinuria/fisiopatología , Ratas Endogámicas Dahl , Ratas Mutantes , Eliminación Renal , Cloruro de Sodio Dietético , Linfocitos T/metabolismo , Factores de Tiempo , Familia-src Quinasas/metabolismo
18.
Physiol Genomics ; 48(6): 409-19, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27113531

RESUMEN

Through linkage analysis of the Dahl salt-sensitive (S) rat and the spontaneously hypertensive rat (SHR), a blood pressure (BP) quantitative trait locus (QTL) was previously located on rat chromosome 9. Subsequent substitution mapping studies of this QTL revealed multiple BP QTLs within the originally identified logarithm of odds plot by linkage analysis. The focus of this study was on a 14.39 Mb region, the distal portion of which remained unmapped in our previous studies. High-resolution substitution mapping for a BP QTL in the setting of a high-salt diet indicated that an SHR-derived congenic segment of 787.9 kb containing the gene secreted phosphoprotein-2 (Spp2) lowered BP and urinary protein excretion. A nonsynonymous G/T polymorphism in the Spp2 gene was detected between the S and S.SHR congenic rats. A survey of 45 strains showed that the T allele was rare, being detected only in some substrains of SHR and WKY. Protein modeling prediction through SWISSPROT indicated that the predicted protein product of this variant was significantly altered. Importantly, in addition to improved cardiovascular and renal function, high salt-fed congenic animals carrying the SHR T variant of Spp2 had significantly lower bone mass and altered bone microarchitecture. Total bone volume and volume of trabecular bone, cortical thickness, and degree of mineralization of cortical bone were all significantly reduced in congenic rats. Our study points to opposing effects of a congenic segment containing the prioritized candidate gene Spp2 on BP and bone mass.


Asunto(s)
Presión Sanguínea/genética , Huesos/metabolismo , Cromosomas Humanos Par 9/genética , Fosfoproteínas/genética , Sitios de Carácter Cuantitativo/genética , Alelos , Animales , Animales Congénicos/genética , Mapeo Cromosómico/métodos , Ligamiento Genético/genética , Humanos , Hipertensión/genética , Masculino , Ratas , Ratas Endogámicas Dahl , Ratas Endogámicas SHR/genética , Ratas Endogámicas WKY , Cloruro de Sodio Dietético/administración & dosificación
19.
J Mol Cell Cardiol ; 88: 101-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26388265

RESUMEN

The transcriptional regulation of pathological cardiac hypertrophy involves the interplay of transcription factors and chromatin remodeling enzymes. The Microphthalmia-Associated Transcription Factor (MITF) is highly expressed in cardiomyocytes and is required for cardiac hypertrophy. However, the transcriptional mechanisms by which MITF promotes cardiac hypertrophy have not been elucidated. In this study, we tested the hypothesis that MITF promotes cardiac hypertrophy by activating transcription of pro-hypertrophy genes through interactions with the SWI/SNF chromatin remodeling complex. In an in vivo model of cardiac hypertrophy, expression of MITF and the BRG1 subunit of the SWI/SNF complex increased coordinately in response to pressure overload. Expression of MITF and BRG1 also increased in vitro when cardiomyocytes were stimulated with angiotensin II or a ß-adrenergic agonist. Both MITF and BRG1 were required to increase cardiomyocyte size and activate expression of hypertrophy markers in response to ß-adrenergic stimulation. We detected physical interactions between MITF and BRG1 in cardiomyocytes and found that they cooperate to regulate expression of a pro-hypertrophic transcription factor, GATA4. Our data show that MITF binds to the E box element in the GATA4 promoter and facilitates recruitment of BRG1. This is associated with enhanced expression of the GATA4 gene as evidenced by increased Histone3 lysine4 tri-methylation (H3K4me3) on the GATA4 promoter. Thus, in hypertrophic cardiomyoctes, MITF is a key transcriptional activator of a pro-hypertrophic gene, GATA4, and this regulation is dependent upon the BRG1 component of the SWI/SNF complex.


Asunto(s)
Cardiomegalia/genética , ADN Helicasas/genética , Factor de Transcripción GATA4/genética , Factor de Transcripción Asociado a Microftalmía/genética , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Factores de Transcripción/genética , Angiotensina II/farmacología , Animales , Aorta/cirugía , Secuencia de Bases , Sitios de Unión , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Línea Celular , Constricción Patológica/complicaciones , Constricción Patológica/cirugía , ADN Helicasas/metabolismo , Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/metabolismo , Datos de Secuencia Molecular , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteínas Nucleares/metabolismo , Cultivo Primario de Células , Unión Proteica , Ratas , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
20.
Nat Commun ; 6: 6252, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25687237

RESUMEN

Genome-wide association studies (GWAS) have prioritized a transcription factor, nuclear receptor 2 family 2 (NR2F2), as being associated with essential hypertension in humans. Here we provide evidence that validates this association and indicates that Nr2f2 is a genetic determinant of blood pressure (BP). Using the zinc-finger nuclease technology, the generation of a targeted Nr2f2-edited rat model is reported. The resulting gene-edited rats have a 15 bp deletion in exon 2 leading to a five-amino-acid deletion in the hinge region of the mutant Nr2f2 protein. Both systolic and diastolic blood pressures of the Nr2f2(mutant) rats are significantly lower than controls. Because the hinge region of Nr2f2 is required for interaction with Friend of Gata2 (Fog2), protein-protein interaction is examined. Interaction of Nr2f2(mutant) protein with Fog2 is greater than that with the wild-type Nr2f2, indicating that the extent of interaction between these two transcription factors critically influences BP.


Asunto(s)
Presión Sanguínea/genética , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/fisiología , Hipertensión/genética , Mutación , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Diástole , Modelos Animales de Enfermedad , Hipertensión Esencial , Exones , Femenino , Eliminación de Gen , Estudio de Asociación del Genoma Completo , Masculino , Mapeo de Interacción de Proteínas , Ratas , Sístole , Factores de Transcripción/metabolismo , Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...