Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Science ; 369(6505): 842-846, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32792398

RESUMEN

How is neuropathic pain regulated in peripheral sensory neurons? Importins are key regulators of nucleocytoplasmic transport. In this study, we found that importin α3 (also known as karyopherin subunit alpha 4) can control pain responsiveness in peripheral sensory neurons in mice. Importin α3 knockout or sensory neuron-specific knockdown in mice reduced responsiveness to diverse noxious stimuli and increased tolerance to neuropathic pain. Importin α3-bound c-Fos and importin α3-deficient neurons were impaired in c-Fos nuclear import. Knockdown or dominant-negative inhibition of c-Fos or c-Jun in sensory neurons reduced neuropathic pain. In silico screens identified drugs that mimic importin α3 deficiency. These drugs attenuated neuropathic pain and reduced c-Fos nuclear localization. Thus, perturbing c-Fos nuclear import by importin α3 in peripheral neurons can promote analgesia.


Asunto(s)
Dolor Crónico/fisiopatología , Neuralgia/fisiopatología , Células Receptoras Sensoriales/fisiología , alfa Carioferinas/fisiología , Transporte Activo de Núcleo Celular , Animales , Benzofenonas/farmacología , Dolor Crónico/genética , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Isoxazoles/farmacología , Ratones , Ratones Endogámicos C57BL , Neuralgia/genética , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción AP-1/metabolismo , alfa Carioferinas/genética
2.
Int J Biol Sci ; 15(6): 1252-1260, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31223284

RESUMEN

CD44 is one of biomarkers of liver cancer stem cells (CSCs). The investigation of mechanism of CD44 translocation helps to uncover new molecular pathways participated in the regulation of various cellular processes in CSCs. In the present study, we observed the translocation of CD44 from cytoplasm to nuclear in the reprogramming process of C3A cells, full-length CD44 presented in the nucleus of liver iCSCs. CD44 was bound with importin ß and transportin 1 in liver iCSCs. Inhibition of importin ß transport leads to reduction of CD44 in the nucleus. Translocation of CD44 is also influenced by importin α. Besides, overexpression of naïve pluripotent genes, KLF2, KLF5, DNMT3L, GBX2, ZFP42, ESRRB and DPPA4 were found in liver iCSCs. Inhibition of CD44 leads to the reduction of these naïve genes. Luciferase and chromatin immunoprecipitation (ChIP) assays further identified nuclear CD44 bound to the promoter regions of naïve genes, KLF2, KLF5, and ESRRB functioned as transcriptional activators in liver iCSCs. Our present work provides new insight into the dynamic states and functions of CD44 in iCSCs.


Asunto(s)
Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/metabolismo , Transporte Activo de Núcleo Celular , Biomarcadores/análisis , Biomarcadores/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/análisis , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , alfa Carioferinas/metabolismo , alfa Carioferinas/fisiología , beta Carioferinas/metabolismo , beta Carioferinas/fisiología
3.
Acta Biochim Biophys Sin (Shanghai) ; 51(3): 285-292, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30883648

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most aggressive cancers worldwide. Identification of the molecular mechanisms underlying the development and progression of HCC is particularly important. Here, we demonstrated the expression pattern, clinical significance, and function of Karyopherin α2 (KPNA2) in HCC. The expression of KPNA2 was upregulated in tumor tissue and negatively associated with the survival time, and a significant correlation between KPNA2 expression and aggressive clinical characteristics was established. Both in vitro and in vivo experiments demonstrated that knockdown of KPNA2 reduced migration and proliferation capacities of HCC cells, while over-expression of KPNA2 increased these malignant characteristics. The analysis of the Cancer Genome Atlas cohorts also reveals that high-KPNA2 expression is associated with poor outcome in multiple cancer types. In addition, gene sets enrichment analysis exhibited cell cycle and DNA replication as the top altered pathways in the high-KPNA2 expression group in HCC and other two cancer types. Overall, this study identified KPNA2 as a potential diagnostic and prognostic biomarker in HCC and other neoplasms, probably by regulating cell cycle and DNA replication.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , alfa Carioferinas/fisiología , Adulto , Anciano , Animales , Carcinoma Hepatocelular/mortalidad , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Humanos , Neoplasias Hepáticas/mortalidad , Ratones , Persona de Mediana Edad , Pronóstico , Regulación hacia Arriba , alfa Carioferinas/genética
4.
Cell Signal ; 44: 103-117, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29331583

RESUMEN

Nucleocytoplasmic shuttling via importins is central to the function of eukaryotic cells and an integral part of the processes that lead to many human diseases. In this study, we addressed the role of α and ß importins in the mechanism of endothelial cell (EC) inflammation and permeability, important pathogenic features of many inflammatory diseases such as acute lung injury and atherosclerosis. RNAi-mediated knockdown of importin α4 or α3 each inhibited NF-κB activation, proinflammatory gene (ICAM-1, VCAM-1, and IL-6) expression, and thereby endothelial adhesivity towards HL-60 cells, upon thrombin challenge. The inhibitory effect of α4 and α3 knockdown was associated with impaired nuclear import and consequently, DNA binding of RelA/p65 subunit of NF-κB and occurred independently of IκBα degradation. Intriguingly, knockdown of importins α4 and α3 also inhibited thrombin-induced RelA/p65 phosphorylation at Ser536, showing a novel role of α importins in regulating transcriptional activity of RelA/p65. Similarly, knockdown of importin ß1, but not ß2, blocked thrombin-induced activation of RelA/p65 and its target genes. In parallel studies, TNFα-mediated inflammatory responses in EC were refractory to knockdown of importins α4, α3 or ß1, indicating a stimulus-specific regulation of RelA/p65 and EC inflammation by these importins. Importantly, α4, α3, or ß1 knockdown also protected against thrombin-induced EC barrier disruption by inhibiting the loss of VE-cadherin at adherens junctions and by regulating actin cytoskeletal rearrangement. These results identify α4, α3 and ß1 as critical mediators of EC inflammation and permeability associated with intravascular coagulation.


Asunto(s)
Inflamación/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , alfa Carioferinas/fisiología , beta Carioferinas/fisiología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad de la Membrana Celular , Células Cultivadas , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Fosforilación , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , alfa Carioferinas/genética , beta Carioferinas/genética
5.
Oncol Rep ; 35(6): 3357-62, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27109484

RESUMEN

Tumor onset and progression are associated with dysfunction of the nuclear transport machinery at the level of import and export receptors. However, the role of Karyopherin α2 (KPNA2) in human tongue squamous cell carcinoma (TSCC) remains unknown. We assessed the proliferation, apoptosis and migration of TSCC CAL-27 cells using wound healing, Transwell and MTT assays, western blotting, electron microscopy and acridine orange/ethidium bromide staining following knockdown of KPNA2. The results revealed the antiproliferative, proapoptotic and anti-migratory effects of KPNA2 silencing on the TSCC CAL-27 cells. Moreover, the knockdown of KPNA2 proved to be accompanied by the upregulation of active caspase-3, cytochrome c, Bax, Bad and decreased expression of Bcl-2, p-Bad and XIAP. KPNA2 activated the caspase-dependent pathway in the CAL-27 cells with upregulation of p53, p21Cip1/Waf1 and p16INK4a. Thus, the present study demonstrated that p53/p21Cip1/Waf1/p16INK4a may be an important pathway involved in the function of KPNA2 in TSCC CAL-27 cells.


Asunto(s)
Apoptosis , Carcinoma de Células Escamosas/patología , Neoplasias de la Lengua/patología , Proteína p53 Supresora de Tumor/fisiología , alfa Carioferinas/fisiología , Caspasas/fisiología , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Humanos , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/análisis
6.
J Proteome Res ; 14(4): 1739-51, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25728791

RESUMEN

Karyopherin alpha 2 (KPNA2) is overexpressed in various human cancers and is associated with cancer invasiveness and poor prognosis. Herein, to understand the essential role of KPNA2 protein complexes in cancer progression, we applied stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative proteomic strategy combined with immunoprecipitation (IP) to investigate the differential KPNA2 protein complexes in lung adenocarcinoma cell lines with different invasiveness potentials. We found that 64 KPNA2-interaction proteins displayed a 2-fold difference in abundance between CL1-5 (high invasiveness) and CL1-0 (low invasiveness) cells. Pathway map analysis revealed that the formation of complexes containing KPNA2 and cytoskeleton-remodeling-related proteins, including actin, beta tubulin, tubulin heterodimers, vimentin, keratin 8, keratin 18, and plectin, was associated with cancer invasiveness. IP demonstrated that the levels of KPNA2-vimentin-pErk complexes were significantly higher in CL1-5 cells than in CL1-0 cells. The KPNA2-vimentin-pErk complex was also up-regulated in the advanced stage compared with the early-stage lung adenocarcinoma tissues. Importantly, the levels of pErk as well as cell migration ability were significantly reduced in KPNA2-knockdown cells; however, migration was restored by treatment with pErk phosphatase inhibitors. Collectively, our results demonstrate the usefulness of a SILAC-based proteomic strategy for identifying invasiveness-associated KPNA2 protein complexes and provide new insight into the KPNA2-mediated modulation of cell migration.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias Pulmonares/metabolismo , Complejos Multiproteicos/metabolismo , Invasividad Neoplásica/fisiopatología , Transducción de Señal/fisiología , alfa Carioferinas/metabolismo , Movimiento Celular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Fosforilación , Proteómica/métodos , Transducción de Señal/genética , Vimentina/metabolismo , alfa Carioferinas/fisiología
7.
Exp Cell Res ; 322(1): 159-67, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24275456

RESUMEN

Pancreatic cancer is an aggressive malignancy and one of the leading causes of cancer deaths. The high mortality rate is mostly due to the lack of appropriate tools for early detection of the disease and a shortage of effective therapies. We have previously shown that karyopherin alpha 7 (KPNA7), the newest member of the alpha karyopherin family of nuclear import receptors, is frequently amplified and overexpressed in pancreatic cancer. Here, we report that KPNA7 expression is absent in practically all normal human adult tissues but elevated in several pancreatic cancer cell lines. Inhibition of KPNA7 expression in AsPC-1 and Hs700T pancreatic cancer cells led to a reduction in cell growth and decreased anchorage independent growth, as well as increased autophagy. The cell growth effects were accompanied by an induction of the cell cycle regulator p21 and a G1 arrest of the cell cycle. Interestingly, the p21 induction was caused by increased mRNA synthesis and not defective nuclear transport. These data strongly demonstrate that KPNA7 silencing inhibits the malignant properties of pancreatic cancer cells in vitro and thereby provide the first evidence on the functional role for KPNA7 in human cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Pancreáticas/patología , alfa Carioferinas/fisiología , Transporte Activo de Núcleo Celular , Adulto , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pancreáticas/genética , Interferencia de ARN/fisiología , ARN Interferente Pequeño/farmacología , Receptores Citoplasmáticos y Nucleares/fisiología , Células Tumorales Cultivadas , alfa Carioferinas/antagonistas & inhibidores
8.
Kidney Int ; 85(3): 624-35, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24284509

RESUMEN

Glomerular endothelial cells (GEnCs) contribute to renal injuries in IgA nephropathy (IgAN). Here we profiled microRNAs (miRNAs) in GEnCs treated with conditioned medium from human mesangial cells in vitro. Levels of miR-223 in GEnCs decreased after incubation with the medium prepared with pIgA from patients with glomerular endothelial proliferation and were also decreased in the glomerular tissues of patients with glomerular endothelial proliferation. Mesangial-derived IL-6 caused miR-223 levels to decrease. The addition of exogenous miR-223 inhibited cell proliferation, ICAM-1 expression, and monocyte adhesion. The NF-κB and STAT3 signaling pathways collaborate during the activation process. MiR-223 mimics inhibited the nuclear localization and DNA binding of p65 and STAT3 but had no effect on the expression of upstream molecules. Instead, importin α4 and α5 (multipurpose nuclear transport receptors), validated as targets of miR-223, were responsible for the nuclear transport of p65 and STAT3. Importin α4 and α5 siRNA inhibited the nuclear localization of p65 and STAT3 and prevented cell proliferation and monocyte adhesion. The level of miR-223 in circulating endothelial cells was decreased and related to the clinical and pathological parameters. Thus, miR-223 downregulation promotes glomerular endothelial cell activation by upregulating importin α4 and α5 in IgAN. Monitoring the level of miR-223 in circulating endothelial cells may provide a noninvasive method for evaluating the severity of IgAN.


Asunto(s)
Glomerulonefritis por IGA/patología , Glomérulos Renales/patología , MicroARNs/fisiología , alfa Carioferinas/fisiología , Adulto , Proliferación Celular , Regulación hacia Abajo , Células Endoteliales/fisiología , Femenino , Humanos , Interleucina-6/farmacología , Masculino , MicroARNs/sangre , FN-kappa B/fisiología , Factor de Transcripción STAT3/fisiología , Regulación hacia Arriba
9.
Cell Death Dis ; 4: e745, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23907459

RESUMEN

Karyopherin alpha 2 (KPNA2), a member of the karyopherin family, has a central role in nucleocytoplasmic transport and is overexpressed in many cancers. Our previous study identified KPNA2 as significantly upregulated in epithelial ovarian carcinoma (EOC), correlating with poor survival of patients. However, the precise mechanism of this effect remains unclear. The aim of the present study was to examine the role of KPNA2 in the proliferation and tumorigenicity of EOC cells, and its clinical significance in tumor progression. Real-time quantitative RT-PCR analysis revealed high expression levels of KPNA2 in 162 out of 191 (84.8%) fresh EOC tissues, which was significantly correlated with International Federation of Gynecology and Obstetrics (FIGO) stage, differentiation, histological type, recurrence, and prognosis of EOC patients. Our results showed that upregulation of KPNA2 expression significantly increased the proliferation and tumorigenicity of EOC cells (EFO-21 and SK-OV3) in vitro and in vivo, by promoting cell growth rate, foci formation, soft agar colony formation, and tumor formation in nude mice. By contrast, knockdown of KPNA2 effectively suppressed the proliferation and tumorigenicity of these EOC cells in vitro and in vivo. Our results also indicated that the molecular mechanisms of the effect of KPNA2 in EOC included promotion of G1/S cell cycle transition through upregulation of c-Myc, enhanced transcriptional activity of c-Myc, activation of Akt activity, suppression of FOXO3a activity, downregulation of cyclin-dependent kinase (CDK) inhibitor p21Cip1 and p27Kip1, and upregulation of CDK regulator cyclin D1. Our results show that KPNA2 has an important role in promoting proliferation and tumorigenicity of EOC, and may represent a novel prognostic biomarker and therapeutic target for this disease.


Asunto(s)
Proliferación Celular , Factores de Transcripción Forkhead/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , alfa Carioferinas/fisiología , Animales , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Factor de Transcripción E2F1/metabolismo , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Análisis Multivariante , Trasplante de Neoplasias , Neoplasias Glandulares y Epiteliales/mortalidad , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Pronóstico , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-myc/genética , Transcripción Genética , Carga Tumoral , Regulación hacia Arriba
10.
Biochim Biophys Acta ; 1833(10): 2348-56, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23773962

RESUMEN

The importin (IMP) superfamily of nuclear transport proteins is essential to key developmental pathways, including in the murine testis where expression of the 6 distinct IMPα proteins is highly dynamic. Present predominantly from the spermatocyte stage onwards, IMPα4 is unique in showing a striking nuclear localization, a property we previously found to be linked to maintenance of pluripotency in embryonic stem cells and to the cellular stress response in cultured cells. Here we examine the role of IMPα4 in vivo for the first time using a novel transgenic mouse model in which we overexpress an IMPα4-EGFP fusion protein from the protamine 1 promoter to recapitulate endogenous testicular germ cell IMPα4 expression in spermatids. IMPα4 overexpression did not affect overall fertility, testis morphology/weight or spermatogenic progression under normal conditions, but conferred significantly (>30%) increased resistance to oxidative stress specifically in the spermatid subpopulation expressing the transgene. Consistent with a cell-specific role for IMPα4 in protecting against oxidative stress, haploid germ cells from IMPα4 null mice were significantly (c. 30%) less resistant to oxidative stress than wild type controls. These results from two unique and complementary mouse models demonstrate a novel protective role for IMPα4 in stress responses specifically within haploid male germline cells, with implications for male fertility and genetic integrity.


Asunto(s)
Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Células Germinativas/metabolismo , Estrés Oxidativo , Espermátides/metabolismo , Espermatogénesis , Testículo/metabolismo , alfa Carioferinas/fisiología , Animales , Southern Blotting , Western Blotting , Diferenciación Celular , Núcleo Celular/metabolismo , Proliferación Celular , ADN/genética , Fertilidad , Citometría de Flujo , Células Germinativas/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Protaminas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Espermátides/citología , Testículo/citología
11.
Oncogene ; 31(16): 2101-14, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21909132

RESUMEN

Tumorigenesis and tumor progression are associated with dysfunction of the nuclear transport machinery at the level of import and export receptors (karyopherins). Recent studies have shown that the nuclear import factor karyopherin-α2 (KPNA2) is a novel prognostic marker for poor prognosis in human breast cancer. Based on the well-defined hallmarks of cancer progression, we performed a detailed in vitro characterization of the phenotypic effects caused by KPNA2 overexpression and KPNA2 silencing in benign and malignant human breast cells. KPNA2 overexpression clearly increased proliferation of MCF7 tumor cells and further led to a reduction of cell-matrix adhesion in benign MCF10A cells, whereas cell migration was significantly increased (P<0.0001) in both tumor models. Remarkably, these individual effects of KPNA2 overexpression on proliferation, cell-matrix adhesion and migration resulted in an increased colony spreading of benign MCF10A breast cells and malignant MCF7 tumor cells (P<0.001), which is a hallmark of cancer progression. Conversely, RNA interference-mediated KPNA2 silencing caused a complete inhibition of MCF7 tumor cell proliferation and migration (P<0.0001). In addition, in these experiments apoptosis was increased (P<0.05) and formation of tumor cell colonies was reduced (P<0.01). Thus, KPNA2 overexpression provoked increased aggressiveness of malignant MCF7 breast tumor cells and induced a shift in benign MCF10A breast cells toward a malignant breast cancer phenotype. In conclusion, we demonstrate for the first time in experimental tumor models that forced KPNA2 expression drives malignant features relevant for breast cancer progression, while its silencing is required for the remission of those progressive phenotypes. This study gives clear evidence that KPNA2 acts as a novel oncogenic factor in human breast cancer, in vitro.


Asunto(s)
Neoplasias de la Mama/genética , Movimiento Celular , alfa Carioferinas/fisiología , Neoplasias de la Mama/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Fenotipo , Interferencia de ARN , Regulación hacia Arriba
12.
J Biol Chem ; 286(17): 15126-31, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21454664

RESUMEN

Snail, a zinc finger-containing transcriptional regulator, migrates into the nucleus where it controls gene expression. We demonstrated previously that importin ß1 directly recognizes the zinc finger domain of Snail and transports it into the nucleus. Here, using in vitro and in vivo assays, we show that importin α, an adaptor protein for importin ß1, negatively regulates the nuclear import of Snail mediated by importin ß1. In vitro binding assays indicated that importin α interacted with the zinc finger domain of Snail to compete with the binding of importin ß1 and that Snail did not form a ternary complex with importin α/importin ß1. Overexpression of importin α in A549 cells reduced the endogenous Snail protein level, which was restored by inhibitors of the proteasome and glycogen synthase kinase 3ß. Furthermore, knockdown of importin α by siRNA treatment increased the endogenous Snail protein level in several cancer cell lines. This study provides a novel regulatory mechanism of the nuclear protein import process by importin α and gives an implication to control Snail activity by inhibiting its nuclear localization.


Asunto(s)
Núcleo Celular/metabolismo , Factores de Transcripción/metabolismo , alfa Carioferinas/fisiología , Transporte Activo de Núcleo Celular , Sitios de Unión , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3 , Glucógeno Sintasa Quinasa 3 beta , Humanos , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Factores de Transcripción de la Familia Snail , Dedos de Zinc , beta Carioferinas/metabolismo
13.
FEBS J ; 278(9): 1561-72, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21371262

RESUMEN

Importin α mediates the nuclear import of proteins through nuclear pore complexes in eukaryotic cells, and is common to all eukaryotes. Previous reports identified at least six importin α family genes in mice. Although these isoforms show differential binding to various import cargoes in vitro, the in vivo physiological roles of these mammalian importin α isoforms remain unknown. Here, we generated and examined importin α5 knockout (impα5(-/-)) mice. These mice developed normally, and showed no gross histological abnormalities in most major organs. However, the ovary and uterus of impα5(-/-) female mice exhibited hypoplasia. Furthermore, we found that impα5(-/-) female mice had a 50% decrease in serum progesterone levels and a 57% decrease in progesterone receptor mRNA levels in the ovary. Additionally, impα5(-/-) uteruses that were treated with exogenous gonadotropins displayed hypertrophy, similarly to progesterone receptor-deficient mice. Although these mutant female mice could become pregnant, the total number of pups was significantly decreased, and some of the pups were dead at birth. These results suggest that importin α5 has essential roles in the mammalian female reproductive organs.


Asunto(s)
Parto Obstétrico , Preñez , alfa Carioferinas/fisiología , Animales , Femenino , Genitales Femeninos/patología , Ratones , Ratones Noqueados , Embarazo , Progesterona/sangre , Receptores de Estrógenos/genética , alfa Carioferinas/genética
14.
Biochim Biophys Acta ; 1813(9): 1562-77, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20977914

RESUMEN

Although proteins are translated on cytoplasmic ribosomes, many of these proteins play essential roles in the nucleus, mediating key cellular processes including but not limited to DNA replication and repair as well as transcription and RNA processing. Thus, understanding how these critical nuclear proteins are accurately targeted to the nucleus is of paramount importance in biology. Interaction and structural studies in the recent years have jointly revealed some general rules on the specificity determinants of the recognition of nuclear targeting signals by their specific receptors, at least for two nuclear import pathways: (i) the classical pathway, which involves the classical nuclear localization sequences (cNLSs) and the receptors importin-α/karyopherin-α and importin-ß/karyopherin-ß1; and (ii) the karyopherin-ß2 pathway, which employs the proline-tyrosine (PY)-NLSs and the receptor transportin-1/karyopherin-ß2. The understanding of specificity rules allows the prediction of protein nuclear localization. We review the current understanding of the molecular determinants of the specificity of nuclear import, focusing on the importin-α•cargo recognition, as well as the currently available databases and predictive tools relevant to nuclear localization. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Señales de Localización Nuclear/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bases de Datos de Proteínas , Humanos , Ratones , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Señales de Localización Nuclear/química , Señales de Localización Nuclear/genética , Proteína Relacionada con la Hormona Paratiroidea/química , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal/fisiología , Proteína 2 de Unión a Elementos Reguladores de Esteroles/química , Proteína 2 de Unión a Elementos Reguladores de Esteroles/fisiología , alfa Carioferinas/química , alfa Carioferinas/fisiología , beta Carioferinas/química , beta Carioferinas/fisiología
16.
Mol Biol Cell ; 20(20): 4412-23, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19710421

RESUMEN

Expression of prohibitin 1 (PHB), a multifunctional protein in the cell, is decreased during inflammatory bowel disease (IBD). Little is known regarding the regulation and role of PHB during intestinal inflammation. We examined the effect of tumor necrosis factor alpha (TNF-alpha), a cytokine that plays a central role in the pathogenesis of IBD, on PHB expression and the effect of sustained PHB expression on TNF-alpha activation of nuclear factor-kappa B (NF-kappaB) and epithelial barrier dysfunction, two hallmarks of intestinal inflammation. We show that TNF-alpha decreased PHB protein and mRNA abundance in intestinal epithelial cells in vitro and in colon mucosa in vivo. Sustained expression of prohibitin in intestinal epithelial cells in vitro and in vivo (prohibitin transgenic mice, PHB TG) resulted in a marked decrease in TNF-alpha-induced nuclear translocation of the NF-kappaB protein p65, NF-kappaB/DNA binding, and NF-kappaB-mediated transcriptional activation despite robust IkappaB-alpha phosphorylation and degradation and increased cytosolic p65. Cells overexpressing PHB were protected from TNF-alpha-induced increased epithelial permeability. Expression of importin alpha3, a protein involved in p50/p65 nuclear import, was decreased in cells overexpressing PHB and in colon mucosa of PHB TG mice. Restoration of importin alpha3 levels sustained NF-kappaB activation by TNF-alpha during PHB transfection. These results suggest that PHB inhibits NF-kappaB nuclear translocation via a novel mechanism involving alteration of importin alpha3 levels. TNF-alpha decreases PHB expression in intestinal epithelial cells and restoration of PHB expression in these cells can protect against the deleterious effects of TNF-alpha and NF-kappaB on barrier function.


Asunto(s)
Proteínas Represoras/fisiología , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , alfa Carioferinas/fisiología , Adenocarcinoma/patología , Animales , Colon/citología , Neoplasias del Colon/patología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Genes Reporteros , Humanos , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prohibitinas , Transporte de Proteínas/fisiología , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras/genética , Factor de Necrosis Tumoral alfa/farmacología
18.
Plant J ; 50(1): 149-58, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17346267

RESUMEN

The receptor importin-alpha mediates the nuclear import of functionally diverse cargo proteins that contain arginine/lysine-rich nuclear localization signals (NLSs). Functional homologs of importin-alpha have been characterized in a wide range of species including yeast, human and plants. However, the differential cargo selectivity of plant importin-alpha homologs has not been established. To advance nuclear import studies conducted in plant cells, we have developed a method that allows importin-alpha-dependent nuclear import to be assayed in Nicotiana benthamiana. We employed virus-induced gene silencing (VIGS) to knock down the expression of two importin-alpha homologs, NbImpalpha1 and NbImpalpha2, which we identified from N. benthamiana. Agro-infiltration was then used to transiently express the NLS-containing proteins Arabidopsis thaliana fibrillarin 1 (AtFib1) and the Nuk6, Nuk7 and Nuk12 candidate effector proteins of the oomycete plant pathogen Phytophthora infestans. In this manner, we demonstrate importin-alpha-dependent nuclear import of Nuk6 and Nuk7. In contrast, the nuclear import of Nuk12 and AtFib1 was unaffected in cells of NbImpalpha-silenced plants. These data suggest that P. infestans Nuk6 and Nuk7 proteins are dependent on one or more alpha-importins for nuclear import. Our VIGS-based assay represents a powerful new technique to study mechanisms underlying the transport of proteins from cytoplasm to nucleus in plants.


Asunto(s)
Núcleo Celular/metabolismo , Nicotiana/metabolismo , Proteínas de Plantas/fisiología , alfa Carioferinas/fisiología , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , ADN Complementario/química , ADN Complementario/genética , Regulación de la Expresión Génica de las Plantas , Silenciador del Gen , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Datos de Secuencia Molecular , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/fisiología , Filogenia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Nicotiana/genética , alfa Carioferinas/genética , alfa Carioferinas/metabolismo
19.
Biochem J ; 403(3): 397-407, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17217336

RESUMEN

Max is a ubiquitous transcription factor with a bHLHZip [basic HLH (helix-loop-helix) leucine zipper] DNA-binding/dimerization domain and the central component of the Myc/Max/Mad transcription factor network that controls cell growth, proliferation, differentiation and apoptotic cell death in metazoans. Max is the obligatory DNA-binding and dimerization partner for all the bHLHZip regulators of the Myc/Max/Mad network, including the Myc family of oncoproteins and the Mad family of Myc antagonists, which recognize E-box DNA elements in the regulatory regions of target genes. Max lacks a transcription regulatory domain and is the only member of the network that efficiently homodimerizes. Binding of Max homodimers to E-box elements suppresses the transcription regulatory functions of its network partners and of other non-network E-box-binding regulators. In contrast with its highly regulated partners, Max is a constitutively expressed and phosphorylated protein. Phosphorylation is, however, the only Max post-translational modification identified so far. In the present study, we have analysed Max posttranslational modifications by MS. We have found that Max is acetylated at several lysine residues (Lys-57, Lys-144 and Lys-145) in mammalian cells. Max acetylation is stimulated by inhibitors of histone deacetylases and by overexpression of the p300 co-activator/HAT (histone acetyltransferase). The p300 HAT also directly acetylates Max in vitro at these three residues. Interestingly, the three Max residues acetylated in vivo and in vitro by p300 are important for Max nuclear localization and Max-mediated suppression of Myc transactivation. These results uncover novel post-translational modifications of Max and suggest the potential regulation of specific Max complexes by p300 and reversible acetylation.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proteínas de Ciclo Celular/metabolismo , Histona Acetiltransferasas/metabolismo , Lisina/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Secuencia de Aminoácidos , Células HeLa , Humanos , Lisina/fisiología , Señales de Localización Nuclear/fisiología , Espectrometría de Masas en Tándem , Transfección , alfa Carioferinas/fisiología , Factores de Transcripción p300-CBP
20.
J Mol Histol ; 37(5-7): 293-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16752129

RESUMEN

During the past 20 years, the MRE11-RAD50-NBS1 complex has become an increasingly important focus in basic and clinical cancer research. One main conceptual step forward was made with the discovery of NBS1 and the understanding of its critical pathophysiological role in Nijmegen breakage syndrome. Major efforts were carried out to define the role in DNA repair of this complex. Recently, basic research has continuously extended our understanding of the complexity of the NBS1 complex. MRE11-RAD50-NBS1 complex can no longer be viewed as having a single role in DNA damage repair since it also serves as a sensor and a mediator in cell cycle checkpoint signaling. Meanwhile, studies have challenged the concept that NBS1 only functions as a tumor suppressor in preserving genome integrity in the nucleus. It may also provide an oncogenic role in the cytoplasm which is associated with the PI3-kinase/AKT-activation pathway. Consistent with this aspect, a growing body of clinical evidence suggests that NBS1 contains a deleterious character that depends on its subcellular localization. This review focuses on recent experimental evidences demonstrating how NBS1 is translocated into the nucleus by an importin KPNA2 which mediates NBS1 subcellular localization and the functions of the NBS1 complex in tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Síndrome de Nijmegen/genética , Proteínas Nucleares/genética , alfa Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de Ciclo Celular/fisiología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Modelos Biológicos , Neoplasias/etiología , Neoplasias/metabolismo , Proteínas Nucleares/fisiología , Transducción de Señal , alfa Carioferinas/genética , alfa Carioferinas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA