Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163298

RESUMEN

Nischarin (Nisch) is a cytosolic scaffolding protein that harbors tumor-suppressor-like characteristics. Previous studies have shown that Nisch functions as a scaffolding protein and regulates multiple biological activities. In the current study, we prepared a complete Nisch knockout model, for the first time, by deletion of exons 5 and 6. This knockout model was confirmed by Qrt-PCR and Western blotting with products from mouse embryonic fibroblast (MEF) cells. Embryos and adult mice of knockouts are significantly smaller than their wild-type counterparts. Deletion of Nisch enhanced cell migration, as demonstrated by wound type and transwell migration assays. Since the animals were small in size, we investigated Nisch's effect on metabolism by conducting several assays using the Seahorse analyzer system. These data indicate that Nisch null cells have lower oxygen consumption rates, lower ATP production, and lower levels of proton leak. We examined the expression of 15 genes involved in lipid and fat metabolism, as well as cell growth, and noted a significant increase in expression for many genes in Nischarin null animals. In summary, our results show that Nischarin plays an important physiological role in metabolic homeostasis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Receptores de Imidazolina/metabolismo , Consumo de Oxígeno/genética , Adenosina Trifosfato/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Respiración de la Célula , Fibroblastos , Expresión Génica/genética , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Consumo de Oxígeno/fisiología
2.
Biochem Biophys Res Commun ; 585: 29-35, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34781058

RESUMEN

Epidemiologic evidence has suggested a relationship between di (2-ethylhexyl) phthalate (DEHP) prenatal exposure and autism spectrum disorders (ASD), but the underlying mechanisms are still at large unknown. In this study, pregnant mice were intragastrically administered with DEHP once a day from GD 3 to GD 17 and the neurobehavioral changes of offspring were evaluated. In addition to the repetitive stereotyped behaviors, DEHP at the concentration of 50 mg/kg/day and above significantly impaired the sociability of the offspring (P < 0.05) and decreased the density of dendritic spines of pyramidal neurons in the prefrontal cortex (P < 0.05). At the same time, the expression of Nischarin protein in prefrontal lobe increased (P < 0.05). Similarly, after 12-h incubation of DEHP at the concentration of 100 nM, the total spine density, especially the mushroom and stubby spine populations, significantly decreased in the primary cultured prefrontal cortical neurons (P < 0.05). However, the inhibitory effect of DEHP were reversed by knockdown of Nischarin expression. Collectively, these results suggest that prenatal DEHP exposure induces Nischarin expression, causes dendritic spine loss, and finally leads to autism-like behavior in mouse offspring.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Dietilhexil Ftalato/toxicidad , Receptores de Imidazolina/metabolismo , Corteza Prefrontal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Trastorno del Espectro Autista/inducido químicamente , Línea Celular Tumoral , Células Cultivadas , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/fisiología , Femenino , Receptores de Imidazolina/genética , Ratones Endogámicos ICR , Plastificantes/toxicidad , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Conducta Social
3.
Cell ; 178(3): 521-535.e23, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31348885

RESUMEN

Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.


Asunto(s)
Benzamidas/metabolismo , Compuestos Bicíclicos con Puentes/farmacología , Heptanos/farmacología , Lisosomas/efectos de los fármacos , Proteínas de Transporte Vesicular/metabolismo , Factor de Transcripción Activador 6/metabolismo , Animales , Benzamidas/química , Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/uso terapéutico , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Mutación del Sistema de Lectura , Heptanos/uso terapéutico , Humanos , Receptores de Imidazolina/antagonistas & inhibidores , Receptores de Imidazolina/genética , Receptores de Imidazolina/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mucina-1/química , Mucina-1/genética , Mucina-1/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas de Transporte Vesicular/química
4.
Cancer Res ; 79(9): 2099-2101, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31043427

RESUMEN

The intercellular exchange of exosomes may play a regulatory function in tumor progression and metastasis. Maziveyi and colleagues demonstrated that Nischarin regulated the secretion of exosomes from breast cancer cells. Loss of Nischarin expression increased exosome production and promoted tumor cell growth and migration, supporting that Nischarin can influence the behavior of surrounding cancer cells. This study identified a novel function of the tumor suppressor Nischarin in exosome biology and cancer progression.See related article by Maziveyi et al., p. 2152.


Asunto(s)
Neoplasias de la Mama/genética , Exosomas , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular
5.
Biochem Biophys Res Commun ; 512(3): 460-466, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30902386

RESUMEN

Morphine is a potent opioid analgesic used to alleviate moderate or severe pain, but the development of drug tolerance and dependence limits its use in pain management. Our previous studies showed that the candidate protein for I1 imidazoline receptor, imidazoline receptor antisera-selected (IRAS)/Nischarin, interacts with µ opioid receptor (MOR) and modulates its trafficking. However, there is no report of the effect of IRAS on morphine tolerance and physical dependence. In the present study, we found that IRAS knockout (KO) mice showed exacerbated analgesic tolerance and physical dependence compared to wild-type (WT) mice by chronic morphine treatment. Chronic morphine treatment down-regulated the expression of MOR in spinal cord of IRAS KO mice, while had no significant effect on MOR expression in WT mice. We observed the compensatory increase of cAMP accumulation in spinal cord after morphine tolerance, and this change was more significant in KO mice than WT mice. Furthermore, KO mice showed more elevation in the phosphorylation of AMPA receptor GluR1-S845 than WT mice, while the total expression of GluR1 remained unchanged after morphine dependence. Altogether, these data suggest that IRAS may play an important role in the development of morphine tolerance and physical dependence in vivo through modulating MOR expression, as well as AMPA GluR1-S845 phosphorylation, which might be one of the mechanisms underlying the development of opiate addiction.


Asunto(s)
Analgésicos Opioides/farmacología , Tolerancia a Medicamentos , Receptores de Imidazolina/metabolismo , Dependencia de Morfina/metabolismo , Morfina/farmacología , Animales , AMP Cíclico/metabolismo , Receptores de Imidazolina/genética , Ratones , Ratones Noqueados , Dependencia de Morfina/genética
6.
Mol Cancer ; 17(1): 21, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29415725

RESUMEN

BACKGROUND: During metastasis, tumor cells move through the tracks of extracellular matrix (ECM). Focal adhesions (FAs) are the protein complexes that link the cell cytoskeleton to the ECM and their presence is necessary for cell attachment. The tumor suppressor Nischarin interacts with a number of signaling proteins such as Integrin α5, PAK1, LIMK1, LKB1, and Rac1 to prevent cancer cell migration. Although previous findings have shown that Nischarin exerts this migratory inhibition by interacting with other proteins, the effects of these interactions on the entire FA machinery are unknown. METHODS: RT-PCR, Western Blotting, invadopodia assays, and immunofluorescence were used to examine FA gene expression and determine whether Nischarin affects cell attachment, as well as the proteins that regulate it. RESULTS: Our data show that Nischarin prevents cell migration and invasion by altering the expression of key focal adhesion proteins. Furthermore, we have found that Nischarin-expressing cells have reduced ability to attach the ECM, which in turn leads to a decrease in invadopodia-mediated matrix degradation. CONCLUSIONS: These experiments demonstrate an important role of Nischarin in regulating cell attachment, which adds to our understanding of the early events of the metastatic process in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Adhesión Celular/genética , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Podosomas/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Receptores de Imidazolina/metabolismo , Integrinas/genética , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Biológicos , Podosomas/metabolismo
7.
Sci Rep ; 7(1): 12496, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970529

RESUMEN

Chronic otitis media with effusion (COME) is the most common cause of hearing loss in children, and known to have high heritability. Mutant mouse models have identified Fbxo11, Evi1, Tgif1, and Nisch as potential risk loci. We recruited children aged 10 and under undergoing surgical treatment for COME from 35 hospitals in the UK, and their nuclear family. We performed association testing with the loci FBXO11, EVI1, TGIF1 and NISCH and sought to replicate significant results in a case-control cohort from Finland. We tested 1296 families (3828 individuals), and found strength of association with the T allele at rs881835 (p = 0.006, OR 1.39) and the G allele at rs1962914 (p = 0.007, OR 1.58) at TGIF1, and the A allele at rs10490302 (p = 0.016, OR 1.17) and the G allele at rs2537742 (p = 0.038, OR 1.16) at FBXO11. Results were not replicated. This study supports smaller studies that have also suggested association of otitis media with polymorphism at FBX011, but this is the first study to report association with the locus TGIF1. Both FBX011 and TGIF1 are involved in TGF-ß signalling, suggesting this pathway may be important in the transition from acute to chronic middle ear inflammation, and a potential molecular target.


Asunto(s)
Proteínas F-Box/genética , Sitios Genéticos , Proteínas de Homeodominio/genética , Otitis Media con Derrame/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Factor de Crecimiento Transformador beta1/genética , Alelos , Animales , Niño , Preescolar , Enfermedad Crónica , Estudios de Cohortes , Modelos Animales de Enfermedad , Proteínas F-Box/metabolismo , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/metabolismo , Humanos , Receptores de Imidazolina/genética , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína del Locus del Complejo MDS1 y EV11/genética , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Masculino , Ratones , Otitis Media con Derrame/metabolismo , Otitis Media con Derrame/patología , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
8.
Cancer Res ; 75(20): 4252-9, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26392073

RESUMEN

Tumor suppressor genes regulate cell growth and prevent spontaneous proliferation that could lead to aberrant tissue function. Deletions and mutations of these genes typically lead to progression through the cell-cycle checkpoints, as well as increased cell migration. Studies of these proteins are important as they may provide potential treatments for breast cancers. In this review, we discuss a comprehensive overview on Nischarin, a novel protein discovered by our laboratory. Nischarin, or imidazoline receptor antisera-selected protein, is a protein involved in a vast number of cellular processes, including neuronal protection and hypotension. The NISCH promoter experiences hypermethylation in several cancers, whereas some highly aggressive breast cancer cells exhibit genomic loss of the NISCH locus. Furthermore, we discuss data illustrating a novel role of Nischarin as a tumor suppressor in breast cancer. Analysis of this new paradigm may shed light on various clinical questions. Finally, the therapeutic potential of Nischarin is discussed.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Encéfalo/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Receptores de Imidazolina/química , Receptores de Imidazolina/genética , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
9.
Mol Cancer Ther ; 14(5): 1202-12, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25724667

RESUMEN

NISCH encodes the imidazoline receptor Nischarin and is a known tumor suppressor in many human malignancies; however, its roles in ovarian cancer are still largely unknown. Here, we aim to investigate the biologic functions of NISCH in ovarian cancer. We found that NISCH was significantly downregulated, which correlated considerably with advanced tumor stage, poor differentiation, lymph node metastasis, and the serous/mucinous subtypes in a panel of ovarian cancer tissues. Moreover, NISCH gene silencing was mainly the product of promoter hypermethylation, which could be reversed by treatment with 5-aza-dC. In vitro, NISCH overexpression suppressed cell proliferation and colony formation by hindering cell-cycle progression, whereas the opposite was observed in NISCH knockdown counterparts. In vivo, abundant NISCH expression hindered the growth of HO8910 xenografts, whereas NISCH knockdown accelerated the growth of SKOV3 xenografts. In addition, NISCH significantly attenuated cell invasion by inhibiting the phosphorylation of FAK and ERK, which could be neutralized by PF-562271 (a FAK/Pyk2 inhibitor). Accordingly, NISCH knockdown xenografts exhibited increased peritoneal/pelvic metastases that were not present in counterparts treated with PF-562271. Furthermore, NISCH expression in primary ovarian cancer cells predicted a cellular resistance to PF-562271. In conclusion, we showed that NISCH was frequently silenced by promoter hypermethylation in human ovarian cancer. NISCH manipulated cellular proliferation and invasion by arresting cell cycle and inhibiting the FAK signal. Our findings revealed the biologic functions of NISCH in ovarian cancer, and might be useful for treating patients with aberrant expression of NISCH.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Receptores de Imidazolina/genética , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN/efectos de los fármacos , Decitabina , Regulación hacia Abajo , Femenino , Humanos , Pérdida de Heterocigocidad , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Transducción de Señal
10.
Mol Med Rep ; 12(1): 77-82, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25695373

RESUMEN

The present study aimed to investigate the expression of Nischarin protein in primary breast cancer (PBC), and to evaluate its role in tumor metastasis. Paired specimens of breast cancer tissues and adjacent normal tissues were surgically obtained from 60 patients with PBC at the Zhejiang Cancer Hospital (Hangzhou, China). Nischarin protein concentrations were determined by an ELISA assay. Breast cancer tissues exhibited a significantly lower concentration of Nischarin (5.86 ± 3.19 ng/ml) compared with that of the adjacent noncancerous tissues (9.25 ± 3.65 ng/ml; P<0.001). Furthermore, cancer tissue from patients with lymph node metastasis had significantly lower levels of Nischarin protein (4.69 ± 2.40 ng/ml) than those of patients without lymph node metastasis (7.04 ± 3.47 ng/ml; P=0.004). There was no significant difference in Nischarin protein expression levels between patients with grade I, II or III PBC (grade I, 5.44 ± 3.57 ng/ml; grade II, 6.42 ± 3.85 ng/ml and grade III, 5.10 ± 1.18 ng/ml; P=0.765). The significant differences in the expression of Nischarin between: i) Cancer tissue and noncancerous tissue and ii) patients with and without lymph node metastasis, suggested that Nischarin may have a significant role in tumor occurrence and metastasis of breast cancer. Nischarin expression may therefore be used as a marker to predict the invasiveness and metastasis of PBC.


Asunto(s)
Neoplasias de la Mama/genética , Receptores de Imidazolina/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Metástasis Linfática/genética , Adulto , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Metástasis Linfática/patología , Persona de Mediana Edad , Estadificación de Neoplasias , ARN Mensajero/biosíntesis
11.
J Biol Chem ; 288(22): 15495-509, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23572524

RESUMEN

Biallelic inactivation of LKB1, a serine/threonine kinase, has been detected in 30% of lung adenocarcinomas, and inhibition of breast tumor growth has been demonstrated. We have identified the tumor suppressor, Nischarin, as a novel binding partner of LKB1. Our mapping analysis shows that the N terminus of Nischarin interacts with amino acids 44-436 of LKB1. Time lapse microscopy and Transwell migration data show that the absence of both Nischarin and LKB1 from an invasive breast cancer cell line (MDA-MB-231) enhances migration as measured by increased distance and speed of migrating cells. Our data suggest that this is a result of elevated PAK1 and LIMK1 phosphorylation. Moreover, the absence of Nischarin and LKB1 increased tumor growth in vivo. Consistent with this, the percentage of S phase cells was increased, as demonstrated by flow cytometry and enhanced cyclin D1. The absence of Nischarin and LKB1 also led to a dramatic increase in the formation of lung metastases. Our studies, for the first time, demonstrate functional interaction between LKB1 and Nischarin to inhibit cell migration and breast tumor progression. Mechanistically, we show that these two proteins together regulate PAK-LIMK-Cofilin and cyclin D1/CDK4 pathways.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Células Epiteliales/metabolismo , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glándulas Mamarias Humanas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Lim/genética , Quinasas Lim/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Proteínas Serina-Treonina Quinasas/genética , Trasplante Heterólogo , Proteínas Supresoras de Tumor/genética
12.
Epigenetics ; 8(4): 383-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23503203

RESUMEN

We have previously identified a putative tumor suppressor gene, NISCH, whose promoter is methylated in lung tumor tissue as well as in plasma obtained from lung cancer patients. NISCH was observed to be more frequently methylated in smoker lung cancer patients than in non-smoker lung cancer patients. Here, we investigated the effect of tobacco smoke exposure on methylation of the NISCH gene. We tested methylation of NISCH after oral keratinocytes were exposed to mainstream and side stream cigarette smoke extract in culture. Methylation of the promoter region of the NISCH gene was also evaluated in plasma obtained from lifetime non-smokers and light smokers (<20 pack/year), with and without lung tumors, and heavy smokers (20+ pack/year) without disease. Promoter methylation of NISCH was tested by quantitative fluorogenic real-time PCR in all samples. Promoter methylation of NISCH occurred after exposure to mainstream tobacco smoke as well as to side stream tobacco smoke in normal oral keratinocyte cell lines. NISCH methylation was also detected in 68% of high-risk, heavy smokers without detectable tumors. Interestingly, in light smokers, NISCH methylation was present in 69% of patients with lung cancer and absent in those without disease. Our pilot study indicates that tobacco smoke induces methylation changes in the NISCH gene promoter before any detectable cancer. Methylation of the NISCH gene was also found in lung cancer patients' plasma samples. After confirming these findings in longitudinally collected plasma samples from high-risk populations (such as heavy smokers), examining patients for hypermethylation of the NISCH gene may aid in identifying those who should undergo additional screening for lung cancer.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Fumar/efectos adversos , Adulto , Anciano , Estudios de Cohortes , Femenino , Genes Supresores de Tumor , Humanos , Receptores de Imidazolina/sangre , Péptidos y Proteínas de Señalización Intracelular/sangre , Queratinocitos/patología , Neoplasias Pulmonares/sangre , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Productos de Tabaco
13.
EMBO J ; 32(5): 713-27, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23386062

RESUMEN

The intracellular pathogenic bacterium Salmonella enterica serovar typhimurium (Salmonella) relies on acidification of the Salmonella-containing vacuole (SCV) for survival inside host cells. The transport and fusion of membrane-bound compartments in a cell is regulated by small GTPases, including Rac and members of the Rab GTPase family, and their effector proteins. However, the role of these components in survival of intracellular pathogens is not completely understood. Here, we identify Nischarin as a novel dual effector that can interact with members of Rac and Rab GTPase (Rab4, Rab14 and Rab9) families at different endosomal compartments. Nischarin interacts with GTP-bound Rab14 and PI(3)P to direct the maturation of early endosomes to Rab9/CD63-containing late endosomes. Nischarin is recruited to the SCV in a Rab14-dependent manner and enhances acidification of the SCV. Depletion of Nischarin or the Nischarin binding partners--Rac1, Rab14 and Rab9 GTPases--reduced the intracellular growth of Salmonella. Thus, interaction of Nischarin with GTPases may regulate maturation and subsequent acidification of vacuoles produced after phagocytosis of pathogens.


Asunto(s)
Endosomas/microbiología , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Vacuolas/microbiología , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Transporte Biológico , Western Blotting , Endosomas/metabolismo , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Receptores de Imidazolina/genética , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/metabolismo , Lisosomas/microbiología , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Salmonella/microbiología , Técnicas del Sistema de Dos Híbridos , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rac/genética
14.
Horm Metab Res ; 44(9): 645-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22674473

RESUMEN

Imidazoline I1-receptors (I1R) are known to regulate blood pressure and rilmenidine, an agonist, is widely used as antihypertensive agent in clinic. However, the role of I1R in feeding behavior is still unclear. In the present study, we used the agonist of I1R to investigate the effect on hyperphagia in streptozotocin (STZ)-induced diabetic mice. Rilmenidine decreased the food intake of STZ-diabetic mice in a dose-dependent manner. The reduction of food intake was abolished by pretreatment with efaroxan at the dose sufficient to block I1R. Intracerebroventricular (icv) administration of rilmenidine into STZ-diabetic mice also significantly reduced hyperphagia, which was reversed by icv administration of efaroxan. In addition, similar results were observed in STZ-diabetic mice, which received chronic treatment with rilmenidine 3 times daily (t.i.d.) for 7 days. Moreover, the hypothalamic neuropeptide Y (NPY) level was reduced by rilmenidine that was also reversed by pretreatment with efaroxan. In conclusion, the obtained results suggest that rilmenidine can decrease food intake in STZ-diabetic mice through an activation of I1R to lower hypothalamic NPY level.


Asunto(s)
Cerebro/metabolismo , Diabetes Mellitus Tipo 1/genética , Hiperfagia/genética , Receptores de Imidazolina/genética , Animales , Benzofuranos/administración & dosificación , Cerebro/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Femenino , Humanos , Hiperfagia/tratamiento farmacológico , Hiperfagia/metabolismo , Hiperfagia/fisiopatología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Imidazoles/administración & dosificación , Receptores de Imidazolina/agonistas , Receptores de Imidazolina/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Neuropéptido Y/metabolismo , Oxazoles/administración & dosificación , Rilmenidina , Estreptozocina/efectos adversos
15.
J Natl Cancer Inst ; 103(20): 1513-28, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-21917605

RESUMEN

BACKGROUND: Nischarin (encoded by NISCH), an α5 integrin-binding protein, has been identified as a regulator of breast cancer cell invasion. We hypothesized that it might be a tumor suppressor and were interested in its regulation. METHODS: We examined nischarin expression in approximately 300 human breast cancer and normal tissues using quantitative polymerase chain reaction and immunohistochemistry. Loss of heterozygosity analysis was performed by examining three microsatellite markers located near the NISCH locus in normal and tumor tissues. We generated derivatives of MDA-MB-231 human metastatic breast cancer cells that overexpressed nischarin and measured tumor growth from these cells as xenografts in mice; metastasis by these cells after tail vein injection; and α5 integrin expression, Rac, and focal adhesion kinase (FAK) signaling using western blotting. We also generated clones of MCF-7 human breast cancer cells in which nischarin expression was silenced and measured tumor growth in mouse xenograft models (n = 5 for all mouse experiments). P values were from two-sided Student t tests in pairwise comparisons. RESULTS: Normal human breast tissue samples had statistically significantly higher expression of nischarin mRNA compared with tumor tissue samples (mean level in normal breast = 50.7 [arbitrary units], in breast tumor = 16.49 [arbitrary units], difference = 34.21, 95% confidence interval [CI] = 11.63 to 56.79, P = .003), and loss of heterozygosity was associated with loss of nischarin expression. MDA-MB-231 cells in which nischarin was overexpressed had statistically significantly reduced tumor growth and metastasis compared with parental MDA-MB-231 cells (mean volume at day 40, control vs nischarin-expressing tumors, 1977 vs 42.27 mm(3), difference = 1935 mm(3), 95% CI = 395 to 3475 mm(3), P = .025). Moreover, MCF-7 tumor xenografts in which nischarin expression was silenced grew statistically significantly faster than parental cells (mean volume at day 63, tumors with scrambled short hairpin RNA [shRNA] vs with nischarin shRNA, 224 vs 1262 mm(3), difference = 1038 mm(3), 95% CI = 899.6 to 1176 mm(3), P < .001). Overexpression of nischarin was associated with decreased α5 integrin expression, FAK phosphorylation, and Rac activation. CONCLUSION: Nischarin may be a novel tumor suppressor that limits breast cancer progression by regulating α5 integrin expression and subsequently α5 integrin-, FAK-, and Rac-mediated signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Receptores de Imidazolina/metabolismo , Integrina alfa5/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Receptores de Imidazolina/genética , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Lim/metabolismo , Pérdida de Heterocigocidad , Ratones , Repeticiones de Microsatélite , Invasividad Neoplásica , Fosforilación , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Transducción de Señal , Análisis de Matrices Tisulares , Trasplante Heterólogo , Regulación hacia Arriba , Quinasas p21 Activadas/metabolismo
16.
Mol Cell Biol ; 28(11): 3742-56, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18332102

RESUMEN

Nischarin is a novel protein that regulates cell migration by inhibiting p21-activated kinase (PAK). LIM kinase (LIMK) is a downstream effector of PAK, and it is known to play an important role in cell invasion. Here we show that nischarin also associates with LIMK to inhibit LIMK activation, cofilin phosphorylation, and LIMK-mediated invasion of breast cancer cells, suggesting that nischarin regulates cell invasion by negative modulation of the LIMK/cofilin pathway. The amino terminus of nischarin binds to the PDZ and kinase domains of LIMK. Although LIMK activation enhances the interaction with nischarin, only phosphorylation of threonine 508 of LIMK is crucial for the interaction. Inhibition of endogenous nischarin expression by RNA interference stimulates breast cancer cell invasion. Also, nischarin small interfering RNA (siRNA) enhances cofilin phosphorylation. In addition, knock-down of nischarin showed branched projection actin structures. Collectively these data indicate that nischarin siRNA may enhance random migration, resulting in stimulation of invasion.


Asunto(s)
Cofilina 1/metabolismo , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Lim/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Lim/genética , Ratones , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
17.
Pharmacol Rep ; 59(6): 789-94, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18195472

RESUMEN

The present study aimed at investigating the influence of imidazoline receptor ligands on the proliferation of PC12 cells and the involvement of the sphingosine-1-phosphate (S1P) signaling system in this effect. In cultured PC12 cells, S1P (0.3-100 nM) and the I(1) imidazoline receptor ligands moxonidine (0.3 and 1 mM), agmatine (1 mM), idazoxan (10-100 microM) and efaroxan (1-100 microM) concentration-dependently reduced protein contents which were used as estimates for cell number. The antiproliferative effects elicited by the compounds were abolished after knock-down of S1P(1), S1P(2) and S1P(3) receptors by RNA interference indicating an involvement of S1P receptors. In conclusion, the present data add further evidence to the recent finding that effects of imidazoline receptor ligands in PC12 cells are mediated by homo- and heterodimers of members of the S1P receptor family.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Receptores de Imidazolina/agonistas , Agmatina/metabolismo , Animales , Antihipertensivos/metabolismo , Benzofuranos/metabolismo , Idazoxan/metabolismo , Imidazoles/metabolismo , Receptores de Imidazolina/genética , Receptores de Imidazolina/fisiología , Ligandos , Lisofosfolípidos/metabolismo , Células PC12 , Interferencia de ARN , Ratas , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/fisiología , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA