Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-39034764

RESUMEN

This study evaluated the effects of different nutrient matrices, with or without phytase supplementation, on growth performance, nutrient digestibility, ileal amino acid (AA) digestibility, and blood inositol in pigs fed a complex diet based on corn-soybean meal. Four hundred newly weaned cross-bred (Landrace × Yorkshire × Duroc) 21-day-old piglets of initial body weight 6.35 ± 1.91 kg were allotted to one of the five dietary treatments: Control (CNT), a corn-soybean-based standard diet; negative control 1 (NC1), a standard diet with reduced available phosphorus (Av.P) (-0.125%), metabolizable energy (ME) (-40 kcal), and crude protein (CP) (-0.3%); NC1 with 500 phytase units per kilogramme (FTU/kg) (N1P5); negative control 2 (NC2), a standard diet with greater reduction of Av.P (-0.150%), ME (-55 kcal), and CP (-0.45%,); and NC2 with 1000 FTU/kg (N2P10). Piglets were housed in a random arrangement based on sex and body weight and data were analyzed as a randomized complete block design using analysis of variance. Results showed that the body weight and average daily gain of the NC2 treatment were lower (p < 0.05) compared to NC2. Gain to feed ratio was greater (p < 0.05) in the CNT and N1P5 treatments compared to the NC1, NC2, and N2P10 treatments. The CP digestibility was higher (p < 0.05) in N1P5 and N2P10 treatments compared to other treatments. Moreover, the digestibility of phosphorus and calcium was higher (p < 0.05) in N1P5 and N2P10 treatments than in CNT, NC1, and NC2 treatments. The digestibility of non-dispensable AA; histidine, isoleucine, leucine, phenylalanine, and valine were increased (p < 0.05) in N1P5 and N2P10 than in CNT, NC1, and NC2 treatments. Nevertheless, the digestibility of dispensable AA, glutamic acid, was higher (p < 0.05) in N1P5 and N2P10 treatments than in CNT, NC1, and NC2 treatments. Blood myo-inositol concentration was higher (p < 0.05) in N1P5 and N2P10 treatments compared to CNT, NC1, and NC2 treatments in phase 2. These results demonstrated enhanced outcomes under conditions of moderate deficiency, whereas more pronounced deficiencies necessitated increased phytase dosages to observe significant improvements. The efficacy of phytase was evident in its ability to elevate average daily gain, gain to feed ratio, phosphorus and calcium, CP, AA, and blood myo-inositol.

2.
J Allergy Clin Immunol ; 149(1): 340-357, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33957165

RESUMEN

BACKGROUND: Diesel exhaust particles (DEPs) are associated with the prevalence and exacerbation of allergic respiratory diseases, including allergic rhinitis and allergic asthma. However, DEP-induced mechanistic pathways promoting upper airway disease and their clinical implications remain unclear. OBJECTIVE: We sought to investigate the mechanisms by which DEP exposure contributes to nasal polyposis using human-derived epithelial cells and a murine nasal polyp (NP) model. METHODS: Gene set enrichment and weighted gene coexpression network analyses were performed. Cytotoxicity, epithelial-to-mesenchymal transition (EMT) markers, and nasal polyposis were assessed. Effects of DEP exposure on EMT were determined using epithelial cells from normal people or patients with chronic rhinosinusitis with or without NPs. BALB/c mice were exposed to DEP through either a nose-only exposure system or nasal instillation, with or without house dust mite, followed by zinc finger E-box-binding homeobox (ZEB)2 small hairpin RNA delivery. RESULTS: Bioinformatics analyses revealed that DEP exposure triggered EMT features in airway epithelial cells. Similarly, DEP-exposed human nasal epithelial cells exhibited EMT characteristics, which were dependent on ZEB2 expression. Human nasal epithelial cells derived from patients with chronic rhinosinusitis presented more prominent EMT features after DEP treatment, when compared with those from control subjects and patients with NPs. Coexposure to DEP and house dust mite synergistically increased the number of NPs, epithelial disruptions, and ZEB2 expression. Most importantly, ZEB2 inhibition prevented DEP-induced EMT, thereby alleviating NP formation in mice. CONCLUSIONS: Our data show that DEP facilitated NP formation, possibly via the promotion of ZEB2-induced EMT. ZEB2 may be a therapeutic target for DEP-induced epithelial damage and related airway diseases, including NPs.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Pólipos Nasales , Emisiones de Vehículos/toxicidad , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Adulto , Anciano , Alérgenos/administración & dosificación , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Enfermedad Crónica , Células Epiteliales/fisiología , Femenino , Humanos , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Pólipos Nasales/genética , Pyroglyphidae/inmunología , ARN Interferente Pequeño/administración & dosificación , Rinitis/genética , Sinusitis/genética , Adulto Joven
3.
J Allergy Clin Immunol ; 148(2): 461-472.e13, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33667477

RESUMEN

BACKGROUND: Bone morphogenetic proteins (BMPs), which are members of the TGF-ß superfamily, regulate bone remodeling by stimulating osteoblasts and osteoclasts. Although the association between osteitis and poor surgical outcomes is well known in patients with chronic rhinosinusitis (CRS), BMPs have not been fully investigated as potential biomarkers for the prognosis of CRS. OBJECTIVE: Our aim was to investigate the role of BMPs in osteitis in patients with CRS with nasal polyps (NPs) (CRSwNPs), as well as associations between BMPs and inflammatory markers in sinonasal tissues from patients with CRSwNP. METHODS: We investigated the expression of 6 BMPs (BMP-2, BMP-4, BMP-6, BMP-7, BMP-9, and BMP-10) and their cellular origins in NPs of human subjects by using immunohistochemistry and ELISA of NP tissues. Exploratory factor analysis was performed to identify associations between BMPs and inflammatory markers. Air-liquid interface cell culture of human nasal epithelial cells was performed to evaluate the induction of the epithelial-mesenchymal transition by BMPs. RESULTS: Of the 6 BMPs studied, BMP-2 and BMP-7 were associated with refractoriness. Only BMP-2 concentrations were higher in patients with severe osteitis and advanced disease extent according to the computed tomography findings. Eosinophils and some macrophages were identified as cellular sources of BMP-2 in immunofluorescence analysis. An in vitro experiment revealed that BMP-2 induced epithelial-mesenchymal transition in air-liquid interface-cultured human nasal epithelial cells, particularly in a TH2 milieu. CONCLUSION: BMP-2 could reflect the pathophysiology of mucosa and bone remodeling and may be a novel biomarker for refractory CRSwNP.


Asunto(s)
Proteína Morfogenética Ósea 2 , Mucosa Nasal , Pólipos Nasales , Rinitis , Sinusitis , Adulto , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 2/inmunología , Proteína Morfogenética Ósea 2/metabolismo , Enfermedad Crónica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Pólipos Nasales/inmunología , Pólipos Nasales/metabolismo , Rinitis/inmunología , Rinitis/metabolismo , Sinusitis/inmunología , Sinusitis/metabolismo
4.
EMBO Rep ; 20(9): e45907, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31359606

RESUMEN

Long-term memory formation is attributed to experience-dependent gene expression. Dynamic changes in histone methylation are essential for the epigenetic regulation of memory consolidation-related genes. Here, we demonstrate that the plant homeodomain finger protein 2 (PHF2) histone demethylase is upregulated in the mouse hippocampus during the experience phase and plays an essential role in memory formation. PHF2 promotes the expression of memory-related genes by epigenetically reinforcing the TrkB-CREB signaling pathway. In behavioral tests, memory formation is enhanced by transgenic overexpression of PHF2 in mice, but is impaired by silencing PHF2 in the hippocampus. Electrophysiological studies reveal that PHF2 elevates field excitatory postsynaptic potential (fEPSP) and NMDA receptor-mediated evoked excitatory postsynaptic current (EPSC) in CA1 pyramidal neurons, suggesting that PHF2 promotes long-term potentiation. This study provides insight into the epigenetic regulation of learning and memory formation, which advances our knowledge to improve memory in patients with degenerative brain diseases.


Asunto(s)
Histona Demetilasas/metabolismo , Proteínas de Homeodominio/metabolismo , Consolidación de la Memoria/fisiología , Animales , Biología Computacional , Epigénesis Genética/genética , Hipocampo/metabolismo , Histona Demetilasas/genética , Proteínas de Homeodominio/genética , Masculino , Espectrometría de Masas , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
Biochem Biophys Res Commun ; 527(3): 646-653, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32423806

RESUMEN

Neutrophil extracellular trap (NET) is one of the first-line defenses against microbes. Under certain circumstances, however, it also plays an aggravating factor in diverse inflammation-related diseases including cancers and vascular diseases. Our aim is to develop a new method to detect NET in cells and tissues using a DNA-specific fluorescence probe CDr15. CDr15 was characterized to be impermeable to the cell membranes and to emit a strong fluorescence in association with extracellular DNAs in NET. Due to these properties, CDr15 was successfully shown to quantify NETs in vitro and to be applicable for real-time monitoring NET formation in PMA-stimulated neutrophils. Even in formaldehyde-fixed tumor specimens, CDr15 could detect NETs spreading around cancer cells. Compared with DAPI and SYTOX DNA dyes, CDr15 showed a lower level of background fluorescence and a higher specificity in NET detection. Based on these results, we propose CDr15 as a novel marker of NET to be applicable in experimental and clinical studies.


Asunto(s)
ADN/análisis , Trampas Extracelulares/química , Colorantes Fluorescentes/análisis , Neutrófilos/ultraestructura , Células Cultivadas , Humanos , Microscopía Fluorescente , Neoplasias/patología
6.
Mol Carcinog ; 58(5): 654-665, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30575123

RESUMEN

Intermittent hypoxia (IH), a characteristic of obstructive sleep apnea, is known to promote cancer progression and aggressiveness in mouse models. However, little is known regarding the effect of IH on cancer initiation. Here, the effect of IH on carcinogenesis was explored in azoxymethane (AOM) and dextran sodium sulfate (DSS)-induced colon cancer models with three different protocols. In the first protocol, two other application time points (early or late initiation of IH) were applied. In the second protocol, mice were divided into only two groups, and then exposed to either N or IH conditions for 14 days. In the third protocol, a pharmacological inhibition study for anti-inflammation (5-aminosalicylate) or anti-oxidative stress (N-acetylcysteine [NAC]) was performed. The number of tumors was significantly higher in the IH-1 than in the N or IH-2 groups. 8-oxo-2'-deoxyguanosine (8-OHdG) levels were higher in tumors of the IH-1 group than in that of the N and IH-2 groups. Gene expression related to reactive oxygen species production was higher in the IH-1 group than in the N and IH-2 groups, and it showed a positive correlation with 8-OHdG levels. Prior to cancer development 8-OHdG levels were already elevated in colonic epithelial regions in the IH group, possibly due to an imbalance between oxidative stress and antioxidant systems. NAC treatment resulted in a significant reduction in the number of tumors in mice exposed to IH. In conclusion, IH promotes carcinogenesis in a chemically-induced colon cancer model where elevated 8-OHdG may contribute to the increased tumor induction.


Asunto(s)
Azoximetano/toxicidad , Carcinogénesis/patología , Colitis/patología , Neoplasias del Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Hipoxia/fisiopatología , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/metabolismo , Carcinógenos/toxicidad , Colitis/inducido químicamente , Colitis/metabolismo , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos
7.
Biochem Biophys Res Commun ; 499(3): 475-481, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29580991

RESUMEN

Hypoxia-inducible factor 1 (HIF-1) is a key player in cellular response to hypoxia. The stability and transcriptional activity of this protein are oxygen-dependently regulated by the prolyl hydroxylases PHD1-3 and the asparaginyl hydroxylase FIH. Recently, ferritin heavy chain (FTH1) has been characterized to reinforce the HIF-1 signaling pathway in an indirect way through the inhibition of PHD activity by depleting the free iron pool in the cytoplasm. In the present study, we addressed the role of FTH1 in the FIH control of HIF-1 activity. Unexpectedly, immunoprecipitation analyses revealed that FTH1 directly interacted with FIH. In an in vitro hydroxylation assay, FTH1 was found to facilitate the FIH-mediated Asn803 hydroxylation in HIF-1α. As expected, FTH1 prevented the recruitment of p300 to HIF-1α through the Asn803 hydroxylation. In luciferase reporter analyses, FTH1 was found to repress the transcriptional activity of HIF-1α in HCT116 cells under either normoxic or hypoxic conditions. Consequently, FTH1 downregulated the expression of the HIF-1 target genes, such as VEGF, CA9 and GLUT1. Our results suggest a new role of FTH1 as a co-regulator for the FIH-mediated oxygen sensing pathway. Since HIF-1α is involved in pathogenesis of diverse hypoxia-associated diseases, we propose that FTH1 be a potential target in developing new therapeutic strategies against such diseases.


Asunto(s)
Ferritinas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxigenasas de Función Mixta/metabolismo , Proteínas Represoras/metabolismo , Asparagina/metabolismo , Hipoxia de la Célula/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hidroxilación , Oxidorreductasas , Unión Proteica , Transcripción Genética
8.
Biochem Biophys Res Commun ; 500(2): 288-295, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29653105

RESUMEN

The final strategies to care patients with end-stage renal fibrosis rely on dialysis and kidney transplantation. Because such treatments are invasive and cause health problems eventually, it is necessary to develop new therapeutic strategies for delaying the disease progress. We here searched for cytokines showing an anti-fibrotic activity in cell-based experiments. Cystatin C (CST3) and Growth differentiation factor 15 (GDF15) were identified to have anti-fibrotic activities in a cytokine array screening. In primary fibroblasts isolated from the mouse kidneys subjected to ureteral obstruction-induced fibrosis, each cytokine induced apoptotic cell death and reduced collagen production. These anti-fibrotic effects were further augmented by co-administration of both cytokines. Mechanistically, CST3 and GDF15 were found to block the TGF-ß receptor and the N-Myc signaling pathways, respectively. In mice with unilateral ureter obstruction, each cytokine and the combination of two cytokines effectively reduced the fibrotic burden in the subjected kidneys. Therefore, we propose that CST3 and GDF15 could be potential candidates for biopharmaceutics to ameliorate renal fibrosis.


Asunto(s)
Cistatina C/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Factor 15 de Diferenciación de Crecimiento/farmacología , Riñón/patología , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno/biosíntesis , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
9.
BMC Cancer ; 18(1): 30, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29301501

RESUMEN

BACKGROUND: Protein neddylation is a post-translational modification by a covalent conjugation with the neural precursor cell expressed, developmentally downregulated 8 (NEDD8). Although this process has been reported to participate in diverse cellular signaling, little is known about its role in cancer cell migration. Given a recent proteomics report showing that NEDD8 is downregulated in prostate cancer tissues versus normal prostate tissues, we tested the possibility that neddylation plays a role in cancer evolution, and then tried to identify target proteins of the neddylation. METHODS: The neddylation process was inhibited by transfecting cancer cells with NEDD8-targeting siRNAs or by treating the cells with a NAE1 inhibitor MLN4924. Cell migration was evaluated by an in vitro wound-healing assay and a Transwell migration assay. His/NEDD8-conjugated proteins were pulled down with nickel-affinity beads under a denaturing condition, and identified by Western blotting. All data were processed using the Microsoft Excel program and analyzed statistically by two-sided, unpaired Student's t-test. RESULTS: Caveolin-1, which plays a critical role in cell migration, was identified to be conjugated with NEDD8. When the neddylation was inhibited, the phosphorylation of caveolin-1 at Tyr14 was augmented in PC3 and U373MG cells, thereby leading to increased cell migration. Such consequences by neddylation inhibition were abolished in the presence of a Src family kinase inhibitor PP2. CONCLUSIONS: NEDD8 seems to inhibit the Src-mediated phosphorylation of caveolin-1 by modifying the structure of caveolin-1 protein, which blocks the migration of cancer cells. Although the neddylation process is currently regarded as an emerging target for cancer therapy, our results suggest the possibility that the inhibition of neddylation could facilitate cancer invasion or metastasis at least in some types of cancers.


Asunto(s)
Caveolina 1/genética , Movimiento Celular/genética , Proteína NEDD8/genética , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Proliferación Celular/genética , Ciclopentanos/farmacología , Humanos , Masculino , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/patología , Proteolisis , Pirimidinas/farmacología , Transducción de Señal/genética , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/genética
10.
Cell Biol Int ; 42(3): 303-312, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29047187

RESUMEN

The KN motif and ankyrin repeat domain-containing protein (KANK) family is involved in actin cytoskeleton organization and cell motility. Compared with other KANK members, the biological function of KANK3 is not clear. Here, we identified KANK3 as a new substrate for the oxygen sensor hypoxia-inducible factor 1-alpha inhibitor (HIF1AN), which hydroxylates HIF-1/2α and other ankyrin repeat domain-containing proteins at asparagine residues. An in vitro hydroxylation assay clearly demonstrated asparaginyl hydroxylation of KANK3 by HIF1AN, and mass spectroscopic analysis revealed that KANK3 is hydroxylated at three asparagine residues within the ankyrin repeat domain. Bioinformatics analysis revealed that KANK3 downregulation is correlated with a poor prognosis in several types of cancers, including hepatocellular carcinoma (HCC). In HCC cells, KANK3 knockdown enhanced cell migration and invasion, while its overexpression inhibited these cell behaviors. Interestingly, such effects of KANK3 were not observed under hypoxic conditions, suggesting oxygen-dependent activity of KANK3. Based on these data, we propose that KANK3 acts as a tumor suppressor to control cancer behavior in an oxygen-dependent manner.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Hepáticas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Repetición de Anquirina , Asparagina/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proteínas Portadoras/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Genes Supresores de Tumor , Células HEK293 , Células Hep G2 , Humanos , Hidroxilación , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Oxigenasas de Función Mixta/genética , Oxígeno/metabolismo , Proteínas Represoras/genética
11.
Mol Cell ; 38(6): 864-78, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20620956

RESUMEN

To survive in hypoxic environments, organisms must be able to cope with redox imbalance and oxygen deficiency. The SIRT1 deacetylase and the HIF-1alpha transcription factor act as redox and oxygen sensors, respectively. Here, we found that SIRT1 binds to HIF-1alpha and deacetylates it at Lys674, which is acetylated by PCAF. By doing so, SIRT1 inactivated HIF-1alpha by blocking p300 recruitment and consequently repressed HIF-1 target genes. During hypoxia, SIRT1 was downregulated due to decreased NAD(+) levels, which allowed the acetylation and activation of HIF-1alpha. Conversely, when the redox change was attenuated by blocking glycolysis, SIRT1 was upregulated, leading to the deacetylation and inactivation of HIF-1alpha even in hypoxia. In addition, we confirmed the SIRT1-HIF-1alpha interaction in hypoxic mouse tissues and observed in vivo that SIRT1 has negative effects on tumor growth and angiogenesis. Our results suggest that crosstalk between oxygen- and redox-responsive signal transducers occurs through the SIRT1-HIF-1alpha interaction.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sirtuina 1/metabolismo , Acetilación , Animales , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neovascularización Patológica/metabolismo , Oxidación-Reducción , Transporte de Proteínas , Trasplante Heterólogo , Factores de Transcripción p300-CBP/metabolismo
12.
Pflugers Arch ; 469(10): 1359-1371, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28534086

RESUMEN

Cardiac neuronal nitric oxide synthase (nNOS) is an important molecule that regulates intracellular Ca2+ homeostasis and contractility of healthy and diseased hearts. Here, we examined the effects of nNOS on fatty acid (FA) regulation of left ventricular (LV) myocyte contraction in sham and angiotensin II (Ang II)-induced hypertensive (HTN) rats. Our results showed that palmitic acid (PA, 100 µM) increased the amplitudes of sarcomere shortening and intracellular ATP in sham but not in HTN despite oxygen consumption rate (OCR) was increased by PA in both groups. Carnitine palmitoyltransferase-1 inhibitor, etomoxir (ETO), reduced OCR and ATP with PA in sham and HTN but prevented PA potentiation of sarcomere shortening only in sham. PA increased nNOS-derived NO only in HTN. Inhibition of nNOS with S-methyl-L-thiocitrulline (SMTC) prevented PA-induced OCR and restored PA potentiation of myocyte contraction in HTN. Mechanistically, PA increased intracellular Ca2+ transient ([Ca2+]i) without changing Ca2+ influx via L-type Ca2+ channel (I-LTCC) and reduced myofilament Ca2+ sensitivity in sham. nNOS inhibition increased [Ca2+]i, I-LTCC and reduced myofilament Ca2+ sensitivity prior to PA supplementation; as such, normalized PA increment of [Ca2+]i. In HTN, PA reduced I-LTCC without affecting [Ca2+]i or myofilament Ca2+ sensitivity. However, PA increased I-LTCC, [Ca2+]i and reduced myofilament Ca2+ sensitivity following nNOS inhibition. Myocardial FA oxidation (18F-fluoro-6-thia-heptadecanoic acid, 18F-FTHA) was comparable between groups, but nNOS inhibition increased it only in HTN. Collectively, PA increases myocyte contraction through stimulating [Ca2+]i and mitochondrial activity in healthy hearts. PA-dependent cardiac inotropy was limited by nNOS in HTN, predominantly due to its modulatory effect on [Ca2+]i handling.


Asunto(s)
Hipertensión/metabolismo , Miocardio/metabolismo , Miofibrillas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Señalización del Calcio/fisiología , Citoplasma/metabolismo , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Ratas Sprague-Dawley
13.
J Allergy Clin Immunol ; 137(1): 87-98.e7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26342525

RESUMEN

BACKGROUND: Nasal polyps (NPs) imply a refractory clinical course in patients with chronic rhinosinusitis (CRS). Previously, we showed that hypoxia-inducible factor (HIF) 1 could mediate nasal polypogenesis through epithelial-to-mesenchymal transition (EMT). Sirtuin 1 (SIRT1), a histone deacetylase, reportedly suppresses the transcriptional activity of HIF-1. Thus we hypothesized that SIRT1 attenuates nasal polyposis by inhibiting HIF-1-induced EMT. OBJECTIVE: We sought to determine the role of SIRT1 in patients with nasal polyposis. METHODS: The effects of SIRT1 on nasal polypogenesis were investigated in previously developed murine models. Immunohistochemistry, immunoblotting, and immunoprecipitation were done to evaluate SIRT1, EMT, and hypoxic markers in human nasal epithelial cells or sinonasal tissues from the mice and the patients with CRS with or without NPs. RESULTS: SIRT1 transgenic mice had significantly fewer mucosal lesions with epithelial disruption and fewer NPs than wild-type (WT) mice. In addition, resveratrol (a SIRT1 activator) treatment suppressed nasal polypogenesis in WT mice; however, sirtinol (a SIRT1 inhibitor) administration increased the polyp burden in SIRT1 transgenic mice. In sinonasal specimens from patients with CRS, SIRT1 was downregulated in the mucosa from patients with polyps compared with levels seen in patients without polyps. SIRT1 overexpression or activation reversed hypoxia-induced EMT in human nasal epithelial cells. The intranasal transfection of a small hairpin SIRT1 lentiviral vector induced more nasal polypoid lesions in SIRT1 transgenic mice. Finally, mucosal extracts from patients with CRS without NPs increased SIRT1 expression in nasal epithelial cells, whereas those from patients with CRS with NPs did not. CONCLUSION: SIRT1 suppressed NP formation, possibly because of inhibition of HIF-1-induced EMT. Thus nasal epithelium SIRT1 might be a therapeutic target for NPs.


Asunto(s)
Transición Epitelial-Mesenquimal , Pólipos Nasales/inmunología , Sirtuina 1/inmunología , Adulto , Anciano , Animales , Línea Celular , ADN/genética , Células Epiteliales/inmunología , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Mucosa Nasal/citología , Ovalbúmina/inmunología , Plásmidos , ARN Interferente Pequeño , Sirtuina 1/genética
14.
Pharmacol Res ; 105: 146-51, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26816087

RESUMEN

The methylation status of lysine residues in histones determines the transcription of surrounding genes by modulating the chromatin architecture. Jumonji domain-containing histone-lysine demethylases (Jmj-KDMs) remove the methyl moiety from lysine residues in histones by utilizing Fe(2+) and α-ketoglutarate. Since genetic alterations in Jmj-KDMs occur in various human cancers, the roles of Jmj-KDMs in cancer development and progression have been investigated, but still controversial. The KDM7 subfamily, which belongs to the Jmj-KDM family, is an emerging class of transcriptional coactivators because its members erase the repressive marks H3K9me2/1, H3K27me2/1, and H4K20 me1. Recently, KDM7C (alternatively named PHF2) was discovered as a new KDM7 member and identified to play a tumor-suppressive role through the reinforcement of p53-driven growth arrest and apoptosis. In this article, we generally reviewed the roles of Jmj-KDMs in human cancers and more discussed the molecular functions and the clinical significances of KDM7C.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Neoplasias/genética , Animales , Epigénesis Genética , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética
15.
J Immunol ; 193(10): 4924-33, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305321

RESUMEN

The general consensus is that immune cells are exposed to physiological hypoxia in vivo (PhyO2, 2-5% P(O2)). However, functional studies of B cells in hypoxic conditions are sparse. Recently, we reported the expression in mouse B cells of TASK-2, a member of pH-sensitive two-pore domain K(+) channels with background activity. In this study, we investigated the response of K(+) channels to sustained PhyO2 (sustained hypoxia [SH], 3% P(O2) for 24 h) in WEHI-231 mouse B cells. SH induced voltage-independent background K(+) conductance (SH-K(bg)) and hyperpolarized the membrane potential. The pH sensitivity and the single-channel conductance of SH-K(bg) were consistent with those of TASK-2. Immunoblotting assay results showed that SH significantly increased plasma membrane expressions of TASK-2. Conversely, SH failed to induce any current following small interfering (si)TASK-2 transfection. Similar hypoxic upregulation of TASK-2 was also observed in splenic primary B cells. Mechanistically, upregulation of TASK-2 by SH was prevented by si hypoxia-inducible factor-1α (HIF-1α) transfection or by YC-1, a pharmacological HIF-1α inhibitor. In addition, TASK-2 current was increased in WEHI-231 cells overexpressed with O2-resistant HIF-1α. Importantly, [Ca(2+)]c increment in response to BCR stimulation was significantly higher in SH-exposed B cells, which was abolished by high K(+)-induced depolarization or by siTASK-2 transfection. The data demonstrate that TASK-2 is upregulated under hypoxia via HIF-1α-dependent manner in B cells. This is functionally important in maintaining the negative membrane potential and providing electrical driving force to control Ca(2+) influx.


Asunto(s)
Linfocitos B/metabolismo , Señalización del Calcio , Calcio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/farmacología , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Hipoxia de la Célula , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Indazoles/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/genética , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
16.
J Allergy Clin Immunol ; 135(6): 1476-85.e7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25725991

RESUMEN

BACKGROUND: Chronic rhinosinusitis (CRS) with nasal polyps (NPs) in Western populations is associated with TH2 cytokine polarization. IL-25, an IL-17 family cytokine, was recently reported to induce TH2-type immune responses and to contribute to several allergic diseases, such as atopic dermatitis and asthma. However, the role of IL-25 in Asian patients with nasal polyposis remains unclear. OBJECTIVE: We sought to determine the role of IL-25 in Asian patients with nasal polyposis and CRS. METHODS: We investigated IL-25 expression and its cellular origins in NPs of human subjects using immunohistochemistry (IHC), quantitative RT-PCR, and ELISA of NP tissues. Correlations between IL-25 expression and expression of other inflammatory markers in NP tissues were also explored. Anti-IL-25 neutralizing antibody was administered in an ovalbumin- and staphylococcal enterotoxin B-induced murine NP model to confirm the function of IL-25 during nasal polypogenesis. RESULTS: IL-25 expression was upregulated in NP mucosa from patients with CRS with NPs compared with uncinate process tissue from control subjects and those with CRS without NPs. Overexpression of epithelial IL-25 was confirmed by using IHC, and double IHC staining showed that tryptase-positive cells were one of the main sources of IL-25 among immune cells. Furthermore, IL-17 receptor B levels were also increased in immune cells of patients with NPs compared with those in control subjects. In NPs IL-25 mRNA expression positively correlated with the expression of several inflammatory markers, including T-box transcription factor, RAR-related orphan receptor C, GATA3, eosinophil cationic protein, TGF-ß1, and TGF-ß2. IL-25 was more abundant in the murine NP model compared with control mice, and similar correlations between IL-25 and inflammatory markers were observed in murine models. Anti-IL-25 treatment reduced the number of polyps, mucosal edema thickness, collagen deposition, and infiltration of inflammatory cells, such as eosinophils and neutrophils. This treatment also inhibited expression of local inflammatory cytokines, such as IL-4 and IFN-γ. Furthermore, expression of CCL11, CXCL2, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1 in the nasal mucosa was suppressed in the anti-IL-25-treated group. CONCLUSION: Our results suggest that IL-25 secreted from the sinonasal epithelia and infiltrating mast cells plays a crucial role in the pathogenesis of CRS with NPs in Asian patients. In addition, our results suggest the novel possibility of treating nasal polyposis with anti-IL-25 therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Expresión Génica/efectos de los fármacos , Interleucina-17/antagonistas & inhibidores , Pólipos Nasales/tratamiento farmacológico , Rinitis/tratamiento farmacológico , Sinusitis/tratamiento farmacológico , Adulto , Animales , Estudios de Casos y Controles , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Proteína Catiónica del Eosinófilo/genética , Proteína Catiónica del Eosinófilo/inmunología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/patología , Femenino , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Expresión Génica/inmunología , Humanos , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Ratones , Persona de Mediana Edad , Pólipos Nasales/complicaciones , Pólipos Nasales/genética , Pólipos Nasales/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/patología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Rinitis/complicaciones , Rinitis/genética , Rinitis/inmunología , Sinusitis/complicaciones , Sinusitis/genética , Sinusitis/inmunología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/inmunología
17.
Br J Cancer ; 113(8): 1186-96, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26448177

RESUMEN

BACKGROUND: The biological significance of FOXO1, a member of the forkhead box O transcription factor family, in gastric cancer (GC) remains unclear. The present study provides direct evidence of the role of FOXO1 in tumour growth and metastasis of GC in relation to human epidermal growth factor receptor 2 (HER2). METHODS: The expressions of FOXO1 and HER2 were modulated in GC cell lines (SNU-638, MKN45, SNU-216 and NCI-N87) by stable transfections. The effects of transfection on GC phenotypes were evaluated in vitro and in animal models. In addition, the relationship between FOXO1 and HER2 was analysed using GC clinical specimens, cell lines and xenografts. RESULTS: FOXO1 silencing in GC cells increased colony formation and mesenchymal transition in vitro, as well as tumour growth and metastasis in nude mice, whereas HER2 silencing induced the opposite results.. Furthermore, an inverse relationship between FOXO1 and HER2 was found in clinical specimens of GC, GC cells and GC xenograft tumours. Although a negative crosstalk between these two molecules was shown, double knockdown of both FOXO1 and HER2 in GC cells revealed that HER2 silencing reversed the FOXO1 shRNA-induced migration and invasion even without the FOXO1 restoration. CONCLUSIONS: Our results indicate that loss of FOXO1 promotes GC growth and metastasis by upregulating HER2 expression and that the HER2 expression is more critical to the induction of GC cell metastasis. The present study provides evidence that the FOXO1/HER2 pathway may regulate GC progression in a subgroup of GC patients.


Asunto(s)
Proliferación Celular/genética , Factores de Transcripción Forkhead/genética , Metástasis de la Neoplasia/genética , Receptor ErbB-2/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Regulación hacia Arriba/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proteína Forkhead Box O1 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/genética
18.
Gastroenterology ; 147(2): 430-442.e8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24846398

RESUMEN

BACKGROUND & AIMS: Immunoglobulin transcription factor 2 (ITF2) was believed to promote neoplastic transformation via activation of ß-catenin. However, ITF2 recently was reported to suppress colon carcinogenesis. We investigated the roles of ITF2 in colorectal cancer cell lines and tumor formation and growth in mice. METHODS: Levels of ITF2, ß-catenin, and c-Myc were measured in 12 human colorectal tumor samples and by immunohistochemistry. ITF2 regulation of ß-catenin and T-cell factor (TCF) were analyzed using luciferase reporter, reverse-transcription quantitative polymerase chain reaction, flow cytometry, and immunoblot analyses. Mice were given subcutaneous injections of human colorectal cancer cell lines that stably express ITF2, small hairpin RNAs to reduce levels of ITF2, or control plasmids; xenograft tumor growth was assessed. Human colorectal carcinoma tissue arrays were used to associate levels of ITF2 expression and clinical outcomes. RESULTS: Levels of ß-catenin, cMyc, and ITF2 were increased in areas of human colon adenomas and carcinomas, compared with nontumor areas of the same tissues. ITF2 levels were reduced and cMyc levels were increased in areas of carcinoma, compared with adenoma. In human colorectal cancer cell lines, activation of the ß-catenin-TCF4 complex and expression of its target genes were regulated negatively by ITF2. ITF2 inhibited formation of the ß-catenin-TCF4 complex by competing with TCF4 for ß-catenin binding. Stable transgenic expression of ITF2 in human colorectal cancer cell lines reduced their proliferation and tumorigenic potential in mice, whereas small hairpin RNA knockdown of ITF2 promoted growth of xenograft tumors in mice. In an analysis of colorectal tumor tissue arrays, loss of ITF2 from colorectal tumor tissues was associated with poor outcomes of patients. A gene set enrichment analysis supported the negative correlation between the level of ITF2 and activity of the ß-catenin-TCF4 complex. CONCLUSIONS: In human colorectal cancer cell lines and tissue samples, ITF2 appears to prevent activation of the ß-catenin-TCF4 complex and transcription of its gene targets. Loss of ITF2 promotes the ability of colorectal cancer cells to form xenograft tumors, and is associated with tumor progression and shorter survival times of patients.


Asunto(s)
Adenocarcinoma/metabolismo , Adenoma/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Neoplasias Colorrectales/metabolismo , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenoma/genética , Adenoma/patología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Regulación hacia Abajo , Retroalimentación Fisiológica , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factor de Transcripción 4 , Factores de Transcripción/genética , Transfección , Carga Tumoral , beta Catenina/genética
19.
Biochem Biophys Res Commun ; 464(4): 1008-1015, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26187670

RESUMEN

The transcriptional factor hypoxia-inducible factor-1α (HIF-1α) is induced under hypoxia and plays crucial roles in cancer progression and angiogenesis. Protein arginine methyltransferases (PRMTs), 11 isoforms of which have been identified so far, modulates the functions of diverse proteins by catalyzing arginine methylation in post-translational level. PRMT9 (alternatively named FBXO11) and PRMT11 (FBXO10) are expected to have the E3 ubiquitin ligase activity through their F-box domains as well as the methyltrasferase activity. Given previous studies examining roles of 8 PRMT isoforms (PRMT1-8) in the HIF-1 signaling pathway, PRMT1 and PRMT5 were demonstrated to regulate HIF-1α expression in opposite ways. We herein examined if FBXO10 and FBXO11 participate in the HIF-1 signaling pathway. Consequently, the siRNA-mediated knockdown of FBXO11 facilitated HIF-1α expression in various cancer cells and HIF-1-driven gene expressions, but the FBXO10 knockdown did not. Mechanistically, FBXO11 was found to inhibit de novo synthesis of HIF-1α protein by destabilizing HIF-1α mRNA. Since a FBXO11 mutant lacking F-box failed to reverse the HIF-1α expression by FBXO11 knockdown, the FBXO11 regulation of HIF-1α may be attributed to the ubiquitination of some proteins controlling HIF-1α mRNA stability. Considering the oncogenic roles of HIF-1α, FBXO11 is suggested to act as a tumor suppressor and also to be a potential target for cancer therapy.


Asunto(s)
Hipoxia de la Célula/fisiología , Proteínas F-Box/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hipoxia de la Célula/genética , Línea Celular Tumoral , Proteínas F-Box/antagonistas & inhibidores , Proteínas F-Box/genética , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células MCF-7 , Biosíntesis de Proteínas , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , Estabilidad del ARN , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
20.
J Korean Med Sci ; 30(10): 1388-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26425034

RESUMEN

Hypoxia-inducible factor 1alpha (HIF-1α), which transactivates a variety of hypoxia-induced genes, is rapidly degraded under nomoxia through the hydroxylation-ubiquitination-proteasome pathway. In this study, we addressed how HIF-1α is stabilized by proteasome inhibitors. The ubiquitin pool was rapidly reduced after proteasome inhibition, followed by the accumulation of non-ubiquitinated HIF-1α. The poly-ubiquitination of HIF-1α was resumed by restoration of free ubiquitin, which suggests that the HIF-1α stabilization under proteasome inhibition is attributed to depletion of the free ubiquitin pool. Ni(2+) and Zn(2+) also stabilized HIF-1α with depletion of the free ubiquitin pool and these effects of metal ions were attenuated by restoration of free ubiquitin. Ni(2+) and Zn(2+) may disturb the recycling of free ubiquitin, as MG132 does. Based on these results, the state of the ubiquitin pool seems to be another critical factor determining the cellular level of HIF-1α.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Células HCT116 , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Leupeptinas/farmacología , Níquel/química , Regulación hacia Arriba , Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA