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

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Genomics ; 113(6): 4136-4148, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34715294

RESUMEN

Hereditary Spastic Paraplegias (HSP) are a group of rare inherited neurological disorders characterized by progressive loss of corticospinal motor-tract function. Numerous patients with HSP remain undiagnosed despite screening for known genetic causes of HSP. Therefore, identification of novel genetic variations related to HSP is needed. In this study, we identified 88 genetic variants in 54 genes from whole-exome data of 82 clinically well-defined Korean HSP families. Fifty-six percent were known HSP genes, and 44% were composed of putative candidate HSP genes involved in the HSPome and originally reported neuron-related genes, not previously diagnosed in HSP patients. Their inheritance modes were 39, de novo; 33, autosomal dominant; and 10, autosomal recessive. Notably, ALDH18A1 showed the second highest frequency. Fourteen known HSP genes were firstly reported in Koreans, with some of their variants being predictive of HSP-causing protein malfunction. SPAST and REEP1 mutants with unknown function induced neurite abnormality. Further, 54 HSP-related genes were closely linked to the HSP progression-related network. Additionally, the genetic spectrum and variation of known HSP genes differed across ethnic groups. These results expand the genetic spectrum for HSP and may contribute to the accurate diagnosis and treatment for rare HSP.


Asunto(s)
Paraplejía Espástica Hereditaria , Pueblo Asiatico , Exoma , Humanos , Proteínas de Transporte de Membrana/genética , Mutación , República de Corea , Paraplejía Espástica Hereditaria/diagnóstico , Paraplejía Espástica Hereditaria/genética , Espastina/genética
2.
Biochem Biophys Res Commun ; 509(3): 680-686, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30616891

RESUMEN

Cisplatin causes acute kidney injury (AKI) through proximal tubular injury. We investigated the protective effect of the adenosine monophosphate protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) against cisplatin-induced AKI. We investigated whether the AMP-kinase activator AICAR ameliorates cisplatin-induced AKI through the JAK/STAT/SOCS pathway. Male Sprague-Dawley (SD) rats were randomly divided into four groups: control, AICAR, cisplatin, and cisplatin + AICAR. As appropriate to their treatment group, the rats were injected with a single dose of cisplatin (7 mg/kg, i.p.). AICAR was administered to the rats at 100 mg/kg i.p. daily. Blood urea nitrogen (BUN) and serum creatinine were measured. Renal damage was analyzed in sections stained with hematoxylin and eosin (H&E). Renal tissues were also examined by immunohistochemistry and western blot for p-AMPK, Kim-1, cleaved caspase 3, and JAK/STAT/SOCS. For in vitro studies, NRK-52E normal rat kidney cells were treated with cisplatin and/or AICAR. By western blot, we confirmed the expression of p-AMPK and the JAK/STAT/SOCS pathway in NRK-52E cells. AICAR was protective against cisplatin-induced acute tubular injury by up-regulating p-AMPK expression in NRK-52E cells. Protein expression levels of JAK2/STAT1 were markedly ameliorated in NRK-52E cells by AICAR. The protective mechanism of AICAR may be associated with suppression of the JAK2/STAT1 pathway and up-regulation of SOCS1, an inhibitor of the JAK2/STAT1 pathway. The present study demonstrates the protective effects of AICAR against cisplatin-induced AKI and shows a new renoprotective mechanism through the JAK2/STAT1/SOCS1 pathway and apoptosis inhibition. This study suggests that activation of the AMPK activator AICAR might ameliorate cisplatin-induced AKI.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Aminoimidazol Carboxamida/análogos & derivados , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Activadores de Enzimas/uso terapéutico , Ribonucleótidos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Aminoimidazol Carboxamida/uso terapéutico , Animales , Línea Celular , Quinasas Janus/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratas Sprague-Dawley , Factores de Transcripción STAT/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
3.
Clin Exp Nephrol ; 23(8): 995-1003, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30955190

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) is a common disease, affecting about 10% of the general population. The genetic component about CKD incidence in Asian population is not well known. The aim of the study is to find the genetic loci associated with incident CKD and to figure out the effect of genetic variation on the development of CKD. METHODS: We conducted a genome-wide association (GWA) study regarding the development of CKD based on two population-based cohorts of Korean Genome Epidemiology Study. 3617 Koreans from two different cohorts, aged 40-49 years without CKD at initial visit, were included in our analysis. We used 2510 individuals in Ansan as the discovery set and another 1107 individuals from Ansung as the replication set. At baseline, members of both cohorts provided information on creatinine, and DNA samples were collected for genotyping. Single nucleotide polymorphisms that surpassed a significance threshold of P < 5 × 10-3 were selected. RESULTS: A total of 281 among 3617 developed CKD during the follow-up period. Incident CKD group was older (P < 0.001), included more female (P < 0.001), and had more hypertension and diabetes (P < 0.001). We identified 12 SNPs that are associated with incident CKD in the GWA study and made genetic risk score using these SNPs. In multiple Cox regression analysis, genetic risk score was still a significant associated factor (HR 1.311, CI 1.201, 1.431, P < 0.001). CONCLUSIONS: We identified several loci highly associated with incident CKD. The findings suggest the need for further investigations on the genetic propensity for incident CKD.


Asunto(s)
Sitios Genéticos , Polimorfismo de Nucleótido Simple , Insuficiencia Renal Crónica/genética , Transcriptoma , Adulto , Pueblo Asiatico/genética , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Incidencia , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Fenotipo , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/etnología , República de Corea/epidemiología , Medición de Riesgo , Factores de Riesgo
4.
J Allergy Clin Immunol ; 134(1): 195-203, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24698324

RESUMEN

BACKGROUND: Perforin-1 (Prf1) is the predominant cytolytic protein secreted by natural killer (NK) cells. For a rapid immune response, resting NK cells contain high Prf1 mRNA concentrations while exhibiting minimal cytotoxicity caused by a blockage of Prf1 protein synthesis, implying that an unknown posttranscriptional regulatory mechanism exists. OBJECTIVE: We sought to determine whether microRNA-150 (miR-150) posttranscriptionally regulates Prf1 translation in both mouse and human NK cells at rest and at various time points after activation. METHODS: Mouse NK cells with a targeted deletion of miR-150 (miR-150(-/-) NK cells), primary human NK cells, and NK92 MI cells were used to investigate the role of miR-150 in NK cells. NK cell cytotoxicity assays and Western blotting proved that activated miR-150(-/-) NK cells expressed upregulated Prf1, augmenting NK cell cytotoxicity. When immunodeficient mice were injected with miR-150(-/-) NK cells, there was a significant reduction in tumor growth and metastasis of B16F10 melanoma. RESULTS: We report that miR-150 binds to 3' untranslated regions of mouse and human Prf1, posttranscriptionally downregulating its expression. Mouse wild-type NK cells displayed downregulated miR-150 expression in response to IL-15, which led to corresponding repression and induction of Prf1 during rest and after IL-15 activation, respectively. CONCLUSION: Our results indicate that miR-150 is a common posttranscriptional regulator for Prf1 in mouse and human NK cells that represses NK cell lytic activity. Thus the therapeutic control of miR-150 in NK cells could enhance NK cell-based immunotherapy against cancer, providing a better clinical outcome.


Asunto(s)
Citotoxicidad Inmunológica/genética , Células Asesinas Naturales/inmunología , MicroARNs/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Regiones no Traducidas 3' , Animales , Sitios de Unión , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Humanos , Interleucina-15/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/trasplante , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , MicroARNs/inmunología , Perforina , Proteínas Citotóxicas Formadoras de Poros/inmunología , Biosíntesis de Proteínas , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia
5.
Biochem Biophys Res Commun ; 438(2): 264-9, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23880345

RESUMEN

The p53 protein plays a central role in cell cycle arrest and apoptosis in response to diverse stress stimuli. Human ecdysoneless (hEcd) is known for its role in stabilizing the p53 protein level and increasing p53-mediated transcription. Here, we report that thioredoxin interacting protein (TXNIP), a member of the tumor suppressor family, interacts with hEcd and decreases MDM2-mediated p53 ubiquitination, leading to p53 stabilization and an increase in p53 activity. The ectopic overexpression of both TXNIP and Ecd increased actinomycin D-mediated cell death in MCF-7 cells, whereas knockdown of TXNIP and Ecd decreased cell death. These results show that TXNIP is a new regulator of the Ecd-MDM2-p53 loop.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Muerte Celular , Dactinomicina/farmacología , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Tiorredoxinas/metabolismo , Transcripción Genética
6.
Blood ; 118(20): 5476-86, 2011 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21960590

RESUMEN

Perforin (Prf1) and granzyme B (GzmB) are essential effector molecules for natural killer (NK)-cell cytotoxicity, but how Prf1 and GzmB expression is regulated during arming of NK cells is poorly defined. We show that human microRNA (miR)-27a* is a negative regulator of NK-cell cytotoxicity by silencing Prf1 and GzmB expression. Human miR-27a* specifically bound to the 3' untranslated regions of Prf1 and GzmB, down-regulating expression in both resting and activated NK cells, and it functioned as a fine-tuner for homeostasis of the net amount of the effector proteins. Consistent with miR-27a* having an inhibitory role, knockdown of miR-27a* in NK cells dramatically increased cytotoxicity in vitro and decreased tumor growth in a human tumor xenograft model. Thus, NK-cell cytotoxicity is regulated, in part, by microRNA, and modulating endogenous miR-27a* levels in NK cells represents a potential immunotherapeutic strategy.


Asunto(s)
Neoplasias del Colon/inmunología , Granzimas/genética , Células Asesinas Naturales/fisiología , MicroARNs/fisiología , Proteínas Citotóxicas Formadoras de Poros/genética , Regiones no Traducidas 3'/genética , Animales , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Neoplasias del Colon/terapia , Femenino , Sangre Fetal/citología , Silenciador del Gen , Terapia Genética/métodos , Humanos , Células Asesinas Naturales/citología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , MicroARNs/farmacología , Perforina , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Immunol ; 280(1): 1-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23246829

RESUMEN

Vitamin-D3 upregulated protein-1 (VDUP1) is a stress response protein. Pseudomonas aeruginosa (P. aeruginosa) infection is a leading cause of death. Mice infected with live P. aeruginosa exhibit significantly decreased VDUP1 expression. However, the function of VDUP1 during P. aeruginosa-induced mouse bacteremic shock is unknown. To address the function of VDUP1 in P. aeruginosa-infected mice, we constructed a bacteremic shock model wherein both wild-type and VDUP1-deficient mice were infected intra-peritoneally with live P. aeruginosa. We found that VDUP1-deficient mice were more resistant to P. aeruginosa-induced bacteremic shock than wild-type mice, as shown by the increased survival, accelerated bacterial clearance and suppression of cytokine overproduction of the VDUP1-deficient mice. VDUP1 promoted the recruitment of neutrophils into the peritoneal cavities of infected mice. VDUP1 impeded the phagocytosis of non-opsonized P. aeruginosa via phosphatidylinositide 3-kinase (PI3K) pathway in macrophages. P. aeruginosa infection induced the generation of reactive oxygen species (ROS), and the increased production of ROS by the peritoneal cells of VDUP1-deficient mice was advantageous in clearing the bacteria. Overall, VDUP1 aggravates bacteremic shock; thus, VDUP1 can be considered a target molecule for the inhibition of P. aeruginosa-induced bacteremic shock.


Asunto(s)
Proteínas Portadoras/fisiología , Peritonitis/fisiopatología , Infecciones por Pseudomonas/fisiopatología , Choque Séptico/fisiopatología , Tiorredoxinas/fisiología , Animales , Apoptosis/fisiología , Proteínas Portadoras/genética , Quimiotaxis de Leucocito/fisiología , Recuento de Colonia Microbiana , Citocinas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/patología , Cavidad Peritoneal/microbiología , Cavidad Peritoneal/patología , Peritonitis/microbiología , Fagocitosis/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/aislamiento & purificación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Organismos Libres de Patógenos Específicos , Bazo/microbiología , Tiorredoxinas/genética
8.
J Immunol ; 185(2): 917-28, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20543098

RESUMEN

NK cells are capable of killing virus-infected or tumor cells and producing IFN-gamma. Resting NK cells, however, have only minimal cytolytic activity and secrete a low level of IFN-gamma. The cytokine IL-15 can promote the expression of effector functions by resting NK cells. In this study, we demonstrate that suppressor of cytokine signaling 2 (SOCS2) has a novel role in IL-15-primed human NK cell function. SOCS2 expression was upregulated in NK cells following stimulation with IL-15. During IL-15-mediated NK cell priming, SOCS2 interacted with phosphorylated proline-rich tyrosine kinase 2 (Pyk2) at tyrosine 402 (p-Pyk2(Tyr402)) and induced the proteasome-mediated degradation of p-Pyk2(Tyr402) via ubiquitination. Knockdown of SOCS2 resulted in the accumulation of p-Pyk2(Tyr402) and blocked NK cell effector functions. In addition, NK cell cytolytic activity and IFN-gamma production were inhibited by overexpression of the wild-type of Pyk2 but not by the overexpression of tyrosine 402 mutant of Pyk2. These results suggest that SOCS2 regulates human NK cell effector functions via control of phosphorylated Pyk2 depending on IL-15 existence.


Asunto(s)
Quinasa 2 de Adhesión Focal/metabolismo , Interleucina-15/farmacología , Células Asesinas Naturales/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Quinasa 2 de Adhesión Focal/genética , Humanos , Recién Nacido , Interferón gamma/metabolismo , Células Jurkat , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Mutación , Fosforilación/efectos de los fármacos , Unión Proteica , Interferencia de ARN , Receptores de Interleucina-15/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/genética , Tirosina/metabolismo
9.
J Immunol ; 183(12): 7931-8, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19933860

RESUMEN

Cross-linking of NK activating receptors activates phospholipase-gamma and subsequently induces diacylglycerol and Ca(2+) as second messengers of signal transduction. Previous studies reported that Ras guanyl nucleotide-releasing protein (RasGRP) 1, which is activated by diacylglycerol and Ca(2+), is crucial for TCR-mediated Ras-ERK activation. We now report that RasGRP1, which can also be detected in human NK cells, plays an essential role in NK cell effector functions. To examine the role of RasGRP1 in NK cell functions, the expression of RasGRP1 was suppressed using RNA interference. Knockdown of RasGRP1 significantly blocked ITAM-dependent cytokine production as well as NK cytotoxicity. Biochemically, RasGRP1-knockdown NK cells showed markedly decreased ability to activate Ras, ERK, and JNK. Activation of the Ras-MAPK pathway was independently shown to be indispensable for NK cell effector functions via the use of specific pharmacological inhibitors. Our results reveal that RasGRP1 is required for the activation of the Ras-MAPK pathway leading to NK cell effector functions. Moreover, our data suggest that RasGRP1 might act as an important bridge between phospholipase-gamma activation and NK cell effector functions via the Ras-MAPK pathway.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Sangre Fetal/citología , Sangre Fetal/inmunología , Sangre Fetal/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Células K562 , Células Asesinas Naturales/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Cordón Umbilical/irrigación sanguínea , Cordón Umbilical/citología , Cordón Umbilical/inmunología
10.
J Immunol ; 183(4): 2495-505, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19625652

RESUMEN

Hematopoietic stem cells (HSCs) are maintained in a quiescent state in bone marrow (BM) niches by intrinsic and extrinsic signals. The mechanisms regulating the quiescence and mobilization of HSCs, however, remain unclear. In this study, we report that the expression of thioredoxin-interacting protein (TXNIP) is decreased during HSC activation. In Txnip(-/-) mice, the long-term reconstituting HSC population is decreased and exhausted, and its capacity to repopulate is rapidly lost. These effects are associated with hyperactive Wnt signaling, an active cell cycle, and reduced p21 expression under conditions of stress. TXNIP deficiency reduced the CXCL12- and osteopontin-mediated interaction between HSCs and the bone marrow, and impaired homing and retention in the osteoblastic niche, resulting in mobilized HSCs. Therefore, we propose that TXNIP is essential for maintaining HSC quiescence and the interaction between HSCs and the BM niche.


Asunto(s)
Proteínas Portadoras/fisiología , Movimiento Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Fase de Descanso del Ciclo Celular/fisiología , Estrés Fisiológico , Tiorredoxinas/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Proteínas Portadoras/genética , Movimiento Celular/genética , Células Cultivadas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fase de Descanso del Ciclo Celular/genética , Transducción de Señal/genética , Estrés Fisiológico/genética , Tiorredoxinas/genética , Factores de Tiempo , Proteína Wnt1/antagonistas & inhibidores , Proteína Wnt1/fisiología
12.
Front Genet ; 11: 590924, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584793

RESUMEN

Lennox-Gastaut syndrome (LGS) is a severe type of childhood-onset epilepsy characterized by multiple types of seizures, specific discharges on electroencephalography, and intellectual disability. Most patients with LGS do not respond well to drug treatment and show poor long-term prognosis. Approximately 30% of patients without brain abnormalities have unidentifiable causes. Therefore, accurate diagnosis and treatment of LGS remain challenging. To identify causative mutations of LGS, we analyzed the whole-exome sequencing data of 17 unrelated Korean families, including patients with LGS and LGS-like epilepsy without brain abnormalities, using the Genome Analysis Toolkit. We identified 14 mutations in 14 genes as causes of LGS or LGS-like epilepsy. 64 percent of the identified genes were reported as LGS or epilepsy-related genes. Many of these variations were novel and considered as pathogenic or likely pathogenic. Network analysis was performed to classify the identified genes into two network clusters: neuronal signal transmission or neuronal development. Additionally, knockdown of two candidate genes with insufficient evidence of neuronal functions, SLC25A39 and TBC1D8, decreased neurite outgrowth and the expression level of MAP2, a neuronal marker. These results expand the spectrum of genetic variations and may aid the diagnosis and management of individuals with LGS.

13.
Am J Reprod Immunol ; 78(2)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28251711

RESUMEN

PROBLEM: Trophoblasts are known to decrease natural killer (NK) cell cytotoxicity. However, little is known about the interaction between trophoblasts and NK cells during pregnancy. Interleukin-15 (IL-15) is essential for priming NK cells and maximizing their effector functions. We investigated whether trophoblasts regulate NK cell activation via IL-15/IL-2 receptor and its signaling pathways. METHOD OF STUDY: Natural killer-92 cells were primed with human first-trimester trophoblast cells (Sw.71) conditioned medium (CM) and co-cultured with K562 cells. Flow cytometry, Western blot analysis, and real-time PCR were performed to assess NK cell cytotoxicity, IL-15/IL-2 receptor expression, phosphorylation of STAT5 and MAPKs, and mRNA expression of IL-15-related genes. RESULTS: Natural killer-92 cells incubated with Sw.71 CM showed reduced cytotoxicity and IL-15-mediated proliferation, and expression of IL-15/IL-2 receptor subunits. STAT5 phosphorylation, EOMES and T-bet mRNA expressions, and ERK/JNK pathways of NK 92 cells were suppressed by Sw.71 CM. Productions of perforin, granzyme B, and IFN-γ were also downregulated. CONCLUSION: Trophoblasts regulate human NK cell functions via suppression of IL-15/IL-2 receptor expression, transcription factors, and ERK/JNK pathways.


Asunto(s)
Células Asesinas Naturales/inmunología , Receptores de Interleucina-15/inmunología , Trofoblastos/inmunología , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Humanos , Interferón gamma/inmunología , Células K562 , MAP Quinasa Quinasa 4/inmunología , Receptores de Interleucina-2/inmunología , Factor de Transcripción STAT5/inmunología , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología
14.
Am J Reprod Immunol ; 78(2)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28328096

RESUMEN

Decidual stromal cells (DSCs) are known to regulate trophoblast invasion via unveiled mechanism yet. And nucleotide-binding oligomerization domain-containing protein 1 (NOD1) may influence on this DSC-trophoblast interaction. We investigated the mechanism underlying the DSC-mediated regulation of trophoblast invasion and the effect of NOD1 on their cross talk. Using human primary DSCs, BeWo cell invasion was measured. Cytokine secretion and MAP kinase signaling were examined in DSCs following treatment with NOD1 agonist, Tri-DAP. DSCs secreted IL-8 and increased trophoblast invasion. Tri-DAP further increased IL-8 secretion from DSCs via JNK pathway and facilitated both MMP-2 production and trophoblast invasion compared with control. Upon cotreatment of IL-8 and anti-IL-8 antibody to BeWo cells, the number of invading trophoblasts and MMP-2 production decreased significantly. These results suggest that IL-8 from DSCs may play a role to increase the invasiveness of trophoblast cells into the decidua via NOD1/JNK pathway.


Asunto(s)
Decidua/metabolismo , Interleucina-8/metabolismo , Trofoblastos/fisiología , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Decidua/citología , Femenino , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Embarazo , Primer Trimestre del Embarazo , Transducción de Señal , Células del Estroma/metabolismo
15.
PLoS One ; 9(10): e108913, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25313504

RESUMEN

Natural killer (NK) cells are innate immune effector cells that protect against cancer and some viral infections. Until recently, most studies have investigated the molecular signatures of human or mouse NK cells to identify genes that are specifically expressed during NK cell development. However, the mechanism regulating NK cell development remains unclear. Here, we report a regulatory network of potential interactions during in vitro differentiation of human NK cells, identified using genome-wide mRNA and miRNA databases through hierarchical clustering analysis, gene ontology analysis and a miRNA target prediction program. The microRNA (miR)-583, which demonstrated the largest ratio change in mature NK cells, was highly correlated with IL2 receptor gamma (IL2Rγ) expression. The overexpression of miR-583 had an inhibitory effect on NK cell differentiation. In a reporter assay, the suppressive effect of miR-583 was ablated by mutating the putative miR-583 binding site of the IL2Rγ 3' UTR. Therefore, we show that miR-583 acts as a negative regulator of NK cell differentiation by silencing IL2Rγ. Additionally, we provide a comprehensive database of genome-wide mRNA and miRNA expression during human NK cell differentiation, offering a better understanding of basic human NK cell biology for the application of human NK cells in immunotherapy.


Asunto(s)
Diferenciación Celular , Regulación hacia Abajo , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Células Asesinas Naturales/metabolismo , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Análisis por Conglomerados , Bases de Datos Genéticas , Sangre Fetal/citología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Subunidad gamma Común de Receptores de Interleucina/antagonistas & inhibidores , Subunidad gamma Común de Receptores de Interleucina/genética , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , MicroARNs/genética , Interferencia de ARN , Alineación de Secuencia
16.
Cell Metab ; 18(1): 75-85, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23823478

RESUMEN

Reactive oxygen species (ROS) are critical determinants of the fate of hematopoietic stem cells (HSCs) and hematopoiesis. Thioredoxin-interacting protein (TXNIP), which is induced by oxidative stress, is a known regulator of intracellular ROS. Txnip(-/-) old mice exhibited elevated ROS levels in hematopoietic cells and showed a reduction in hematopoietic cell population. Loss of TXNIP led to a dramatic reduction of mouse survival under oxidative stress. TXNIP directly regulated p53 protein by interfering with p53- mouse double minute 2 (MDM2) interactions and increasing p53 transcriptional activity. Txnip(-/-) mice showed downregulation of the antioxidant genes induced by p53. Introduction of TXNIP or p53 into Txnip(-/-) bone marrow cells rescued the HSC frequency and greatly increased survival in mice following oxidative stress. Overall, these data indicate that TXNIP is a regulator of p53 and plays a pivotal role in the maintenance of the hematopoietic cells by regulating intracellular ROS during oxidative stress.


Asunto(s)
Proteínas Portadoras/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Tiorredoxinas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Antioxidantes/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Proteínas Portadoras/genética , Células Cultivadas , Células Madre Hematopoyéticas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/genética , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
17.
Immunol Lett ; 137(1-2): 70-7, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21354208

RESUMEN

Natural killer (NK) cells are differentiated from hematopoietic stem cells (HSCs) which are located at the lowest end of an oxygen gradient within the bone marrow (BM). In this report, we investigated whether oxygen tension could affect NK cell differentiation from hematopoietic cells in vitro. We found that hypoxia led to an inhibition of differentiation in NK cells, and increased oxygen supply alleviated this inhibition and restored NK cell differentiation under hypoxic condition. Hypoxia-treated cells demonstrated reduced mRNA expression of transcription factors (TFs) that have important roles in NK cell differentiation, such as EOMES, T-bet, GATA-3 and ETS-1. Moreover, hypoxia-pretreated cells recovered mRNA expression of TFs when the oxygen tension was changed to normoxia. Our findings suggest that oxygen tension modulates in vitro differentiation of NK cells through the regulation of TF expression.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Oxígeno/administración & dosificación , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteínas de Dominio T Box/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Factor de Transcripción GATA3/genética , Células Madre Hematopoyéticas/fisiología , Humanos , Hipoxia/genética , Hipoxia/patología , Hipoxia/prevención & control , Células Asesinas Naturales/patología , Células Asesinas Naturales/fisiología , Oxígeno/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Proteínas de Dominio T Box/genética
18.
Immunol Lett ; 136(1): 29-36, 2011 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-21126536

RESUMEN

Natural killer (NK) cells act important roles in innate immunity and adaptive immunity. However, the mechanisms governing NK cell development have not been clearly elucidated. Previous studies have shown that an HMG (high-mobility group) protein, TOX, is important for regulating the differentiation program of developing T cells in mice. In this study, we examined the role of TOX in differentiation of human NK cells. Knockdown of TOX in differentiating cells decreased the NK cell population identified by expression of NK surface markers and receptors. In addition, over-expression of TOX enhanced the differentiation of NK cells which give rise to a population showing effector functions of mature NK cells. Moreover, TOX influenced expression of T-bet (T-box expressed in T cells, also as known as Tbx21) during NK cell development. Overall, these results suggest that TOX is required for IL-15-mediated NK cell differentiation and affected expression of T-bet that plays critical roles in NK differentiation and maturation.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/inmunología , Proteínas del Grupo de Alta Movilidad/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Células K562 , ARN Interferente Pequeño/genética , Transcripción Genética
19.
Immunol Lett ; 141(1): 61-7, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21835206

RESUMEN

The IL-22 NKp46(+) innate lymphoid cells, NCR22 cells, are very important for the early host defense against microbial pathogens. We show here that NCR22 cells were differentiated from Lin(-)CD127(+)CD117(+) cells that were derived from hematopoietic precursor cells (HPCs) of mouse bone marrow cells. The combination of low concentrations of IL-23 and IL-15 induced differentiation of NCR22 cells from Lin(-)CD127(+)CD117(+) cells. NCR22 cells expressed a large amount of IL-22 and RORγt, and they had poor cytolytic activity and produced little IFN-γ. Lin(-)CD127(+)CD117(+) cells were very similar to intestinal lamina propria LTi-like cells; both cells dominantly expressed RORγt and IL-22. Meanwhile, Lin(-)CD127(-)CD117(+) cells that were also derived from HPCs did not express RORγt and IL-22, and they developed into conventional NK cells, not into NCR22 cells. These findings revealed that NCR22 cells can be differentiated from Lin(-)CD127(+)CD117(+) cells which are derived from HPCs.


Asunto(s)
Antígenos Ly/metabolismo , Diferenciación Celular/fisiología , Interleucinas/biosíntesis , Células Asesinas Naturales , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Animales , Femenino , Células Madre Hematopoyéticas/metabolismo , Interleucina-15/inmunología , Interleucina-15/metabolismo , Interleucina-23/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Mucosa Intestinal/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Linfocitos/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/metabolismo , Interleucina-22
20.
Int Immunopharmacol ; 10(4): 481-6, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20116458

RESUMEN

NK cells play crucial roles in innate immunity and adaptive immunity. The detailed mechanisms, however, governing NK cell development remains unclear. In this study, we report that YC-1 significantly enhances NK cell populations differentiated from human umbilical cord blood hematopoietic stem cells (HSCs). NK cells increased by YC-1 display both phenotypic and functional features of fully mature NK (mNK) cells, but YC-1 does not affect the activation of mNK cells. YC-1 did not affect cGMP production and phosphorylation of STAT-5 which is essential for IL-15R signaling. On the other hand, YC-1 increased p38 MAPK phosphorylation during NK cell differentiation. Furthermore, p38 inhibitor SB203580 inhibited the differentiation of NK cells enhanced by YC-1. Taken together, these data suggest that YC-1 enhances NK cell differentiation through the activation of p38 MAPK which is involved in NK cell differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Activadores de Enzimas/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Indazoles/farmacología , Células Asesinas Naturales/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , GMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Sangre Fetal/citología , Citometría de Flujo , Humanos , Imidazoles/farmacología , Indazoles/antagonistas & inhibidores , Fosforilación , Piridinas/farmacología , Receptores de Interleucina-15/efectos de los fármacos , Factor de Transcripción STAT5/biosíntesis , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA