Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 286(34): 30097-106, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21705324

RESUMEN

Proper assembly of mitotic spindles requires Hice1, a spindle-associated protein. Hice1 possesses direct microtubule binding activity at its N-terminal region and contributes to intraspindle microtubule nucleation as a subunit of the Augmin complex. However, whether microtubule binding activity of Hice1 is modulated by mitotic regulators remains unexplored. Here, we found that Aurora-A kinase, a major mitotic kinase, specifically binds to and phosphorylates Hice1. We identified four serine/threonine clusters on Hice1 that can be phosphorylated by Aurora-A in vitro. Of the four clusters, the Ser/Thr-17-21 cluster was the most critical for bipolar spindle assembly, whereas other phospho-deficient point mutants had a minimal effect on spindle assembly. Immunostaining with a phospho-Ser-19/20 phospho-specific antibody revealed that phosphorylated Hice1 primarily localizes to spindle poles during prophase to metaphase but gradually diminishes after anaphase. Consistently, the phospho-mimic 17-21E mutant reduced microtubule binding activity in vitro and diminished localization to spindles in vivo. Furthermore, expression of the 17-21E mutant led to decreased association of Fam29a, an Augmin component, with spindles. On the other hand, expression of the phospho-deficient 17-21A mutant permitted intraspindle nucleation but delayed the separation of early mitotic spindle poles and the timely mitotic progression. Taken together, these results suggest that Aurora-A modulates the microtubule binding activity of Hice1 in a spatiotemporal manner for proper bipolar spindle assembly.


Asunto(s)
Metafase/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Profase/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/metabolismo , Aurora Quinasas , Línea Celular Tumoral , Humanos , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Mutación , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Huso Acromático/genética
2.
Front Genet ; 13: 661348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35783262

RESUMEN

Colorectal cancer is one of the most common malignant tumors in the digestive system. Traditional diagnosis and treatment methods have not significantly improved the overall survival of patients. In this study, we explored the value of ATP2A1 as a biomarker in predicting the prognosis of colorectal cancer patients. We used the TCGA database to reveal the relationship between ATP2A1 mRNA level and prognosis, methylation, and immune invasion in colorectal cancer. The results showed that the expression of ATP2A1 was increased in colorectal cancer. The overall survival of patients with high expression of ATP2A1 was significantly lower than patients with low expression of ATP2A1. Cox regression analysis showed that high expression of ATP2A1 was an independent risk factor for poor prognosis in colorectal cancer patients. In addition, we used three datasets to perform a meta-analysis, which further confirmed the reliability of the results. Furthermore, we revealed that ATP2A1 could significantly inhibit the proliferation of colorectal cancer cells by inhibiting the autophagy process and was associated with several immune cells, especially CD8 + T cells. Finally, four small molecule drugs with potential inhibition of ATP2A1 expression were found by CMap analysis. This study demonstrates for the first time that ATP2A1 is a potential pathogenic factor, which may play a significant role in colorectal cancer.

3.
BMC Med Genomics ; 15(1): 180, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35971121

RESUMEN

BACKGROUND: Serine and arginine-rich splicing factor 9 (SRSF9) has been linked to the occurrence and progression of various cancers; however, its effects and mechanism of action hepatocellular carcinoma (HCC) have not been reported. In this study, we used a bioinformatics approach and in vitro assays to evaluate the expression of SRSF9 in HCC, its prognostic value, and its underlying regulatory mechanisms, including analyses of related pathways and the role of methylation. METHODS: Transcriptomic and DNA methylation data for 357 HCC cases and 50 paratumor tissues in The Cancer Genome Atlas database were obtained. Additionally, protein expression data for cell lines and tissue samples were obtained from the Human Protein Atlas. The CMap databased was used to predict candidate drugs targeting SRSF9. Various cell lines were used for in vitro validation. RESULTS: SRSF9 expression was significantly elevated in HCC and was negatively regulated by its methylation site cg06116271. The low expression of SRSF9 and hypermethylation of cg06116271 were both associated with a longer overall survival time. A correlation analysis revealed ten genes that were co-expressed with SRSF9; levels of CDK4, RAN, DENR, RNF34, and ANAPC5 were positively correlated and levels of RBP4, APOC1, MASP2, HP, and HPX were negatively correlated with SRSF9 expression. The knockdown of SRSF9 in vitro inhibited the proliferation and migration of HCC cells and significantly reduced the expression of proteins in the Wnt signaling pathway (DVL2 and ß-catenin) and cell cycle pathway (Cyclin D and Cyclin E). A CMap analysis identified two drugs, camptothecin and apigenin, able to target and inhibit the expression of SRSF9. CONCLUSIONS: This study expands our understanding of the molecular biological functions of SRSF9 and cg06116271 and provides candidate diagnostic and therapeutic targets for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Factores de Empalme Serina-Arginina , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/patología , Proteínas Portadoras , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Factor IX/genética , Factor IX/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/patología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Pronóstico , Factores de Empalme de ARN/genética , Proteínas Plasmáticas de Unión al Retinol , Serina/genética , Serina/metabolismo , Factores de Empalme Serina-Arginina/genética
4.
Mol Cell Biol ; 27(13): 4905-16, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17470549

RESUMEN

Faithful mitotic partitioning of the Golgi apparatus and the centrosome is critical for proper cell division. Although these two cytoplasmic organelles are probably coordinated during cell division, supporting evidence of this coordination is still largely lacking. Here, we show that the RAD50-interacting protein, RINT-1, is localized at the Golgi apparatus and the centrosome in addition to the endoplasmic reticulum. To examine the biological roles of RINT-1, we found that the homozygous deletion of Rint-1 caused early embryonic lethality at embryonic day 5 (E5) to E6 and the failure of blastocyst outgrowth ex vivo. About 81% of the Rint-1 heterozygotes succumbed to multiple tumor formation with haploinsufficiency during their average life span of 24 months. To pinpoint the cellular function of RINT-1, we found that RINT-1 depletion by RNA interference led to the loss of the pericentriolar positioning and dispersal of the Golgi apparatus and concurrent centrosome amplification during the interphase. Upon mitotic entry, RINT-1-deficient cells exhibited multiple abnormalities, including aberrant Golgi dynamics during early mitosis and defective reassembly at telophase, increased formation of multiple spindle poles, and frequent chromosome missegregation. Mitotic cells often underwent cell death in part due to the overwhelming cellular defects. Taken together, these findings suggest that RINT-1 serves as a novel tumor suppressor essential for maintaining the dynamic integrity of the Golgi apparatus and the centrosome, a prerequisite to their proper coordination during cell division.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Aparato de Golgi/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Muerte Celular , División Celular , Supervivencia Celular , Segregación Cromosómica , Cromosomas Humanos/genética , Desarrollo Embrionario , Retículo Endoplásmico/metabolismo , Eliminación de Gen , Células HeLa , Heterocigoto , Homocigoto , Humanos , Interfase , Ratones , Neoplasias/patología , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo
5.
J Org Chem ; 74(5): 2018-27, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19191556

RESUMEN

Two stereoselective routes were developed to synthesize optically pure IBR2 analogues 1-16. The first features addition of N-Boc-3-bromoindole 26 to the sulfinamide 25, providing a 1:1 ratio of the separable diasteroisomers 27 and 28 in good yield. In a straightforward fashion, the sulfinamides 27 and 28 were conveniently converted into the key amines 39 and 47 over 8 steps, respectively, from which a series of 3,4-dihydroisoquinolinyl IBR2 analogues 1-14 containing fluorinated and trifluoromethylated benzyl groups were prepared. Another route highlights the highly enantioselective addition of indole to the sulfonyl amide 50 with bifunctional aminothioureas 57 and 58 as catalysts. After the reaction conditions were optimized, the desired sulfonyl amides (R)-55 and (S)-55 were obtained in 99% ee and 98% ee, respectively. Acylation of (R)-55 and (S)-55 separately and subsequent allylation gave compounds 60 and 63, respectively, which were further subjected to RCM to furnish compounds 61 and 64 and, after removal of the Boc groups, the desired IBR2 analogues 15 and 16.


Asunto(s)
Amidas/química , Aminas/química , Indoles/síntesis química , Tetrahidroisoquinolinas/síntesis química , Aminas/síntesis química , Cristalografía por Rayos X , Indoles/química , Modelos Moleculares , Conformación Molecular , Estereoisomerismo , Tetrahidroisoquinolinas/química
6.
Biochem Biophys Res Commun ; 372(3): 454-8, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18502200

RESUMEN

Human Kif4A is a member of the Kinesin-4 family of kinesins. Kif4A is thought to be a bona fide chromokinesin because it possesses a motor domain and associates with condensed chromosomes during mitosis. Genetic deletion of Kif4A promotes tumorigenic phenotypes in mouse embryonic cells. Kif4A is critical for mitotic regulation including chromosome condensation, spindle organization and cytokinesis. However, the precise chromatin-binding domain of Kif4A has not been characterized. Herein, we report the identification of two conserved motifs critical for chromatin-binding: the first leucine Zip motif (Zip1) of a leucine Zip/Basic/leucine Zip region (ZBZ) previously thought to be a nuclear localization signal (NLS), and a cysteine-rich (CR) motif within the C-terminal region of Kif4A. Furthermore, by depleting endogenous Kif4A via RNAi and concurrently expressing RNAi-resistant Kif4A versions, we observed that wild type Kif4A, but not the mutants deficient in DNA-binding (Zip1 or CR deleted) or ATPase activity (K94A point mutant), was able to rescue the RNAi-elicited abnormal mitotic profile. Taken together, our results show that both the Zip1 and CR motifs are important for Kif4A chromatin-binding and its mitotic function.


Asunto(s)
Cromatina/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Mitosis , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Células HeLa , Humanos , Cinesinas/genética , Ratones , Datos de Secuencia Molecular , Señales de Localización Nuclear , Interferencia de ARN
7.
Lab Chip ; 18(24): 3733-3749, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30397689

RESUMEN

Adoptive T cell transfer, in particular TCR T cell therapy, holds great promise for cancer immunotherapy with encouraging clinical results. However, finding the right TCR T cell clone is a tedious, time-consuming, and costly process. Thus, there is a critical need for single cell technologies to conduct fast and multiplexed functional analyses followed by recovery of the clone of interest. Here, we use droplet microfluidics for functional screening and real-time monitoring of single TCR T cell activation upon recognition of target tumor cells. Notably, our platform includes a tracking system for each clone as well as a sorting procedure with 100% specificity validated by downstream single cell reverse-transcription PCR and sequencing of TCR chains. Our TCR screening prototype will facilitate immunotherapeutic screening and development of T cell therapies.


Asunto(s)
Técnicas Analíticas Microfluídicas/instrumentación , Receptores de Antígenos de Linfocitos T , Análisis de la Célula Individual , Linfocitos T/química , Linfocitos T/citología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Diseño de Equipo , Humanos , Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/análisis , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos , Linfocitos T/metabolismo , Linfocitos T/trasplante
8.
Cancer Res ; 63(10): 2589-95, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12750284

RESUMEN

Immortalized cells maintain telomere length through either a telomerase-dependent process or a telomerase-independent pathway termed alternative lengthening of telomeres (ALT). Homologous recombination is implicated in the ALT pathway in both yeast and human ALT cells. In ALT cells, two types of DNA double-strand break repair and homologous recombination factors, the Rad50/Mre11/NBS1 complex and Rad51/Rad52 along with replication factors (RPA) and telomere binding proteins (TRF1 and TRF2), are associated with the ALT-associated PML body (APB). DNA synthesis in late S-G(2) is associated with APBs, which contain telomeric DNA and, are therefore, potential sites for telomere length maintenance. Here, we show that the breast cancer susceptibility gene product, breast cancer susceptibility gene 1, and the human homologue of yeast Rap1, hRap1, are also associated with APBs specifically during late S-G(2) phase of the cell cycle. We additionally show that the localization of the double-strand break repair factors with APBs is distinct from their association with ionizing radiation-induced nuclear foci. To systematically explore the mechanism involved in the assembly of APBs, we examine the role of Nijmegen breakage syndrome 1 (NBS1) and TRF1 in this process, respectively. We demonstrated that NBS1 plays a key role in the assembly and/or recruitment of Rad50, Mre11, and breast cancer susceptibility gene 1, but not Rad51 or TRF1, to APBs. The NH(2) terminus of NBS1, specifically the BRCA1 COOH-terminal domain, is required for this activity. Although TRF1 interacts with NBS1 directly, it is dispensable for the association of either Rad50/Mre11/NBS1 or Rad51 with APBs. Perturbation of the interactions between NBS1/Mre11 and APBs correlates with reduced BrdUrd incorporation associated with APBs, consistent with decreased DNA synthesis at these sites. Taken together, these results support a model in which NBS1 has a vital role in the assembly of APBs, which function to maintain telomeres in human ALT cells.


Asunto(s)
Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Enzimas Reparadoras del ADN , Proteínas Nucleares/metabolismo , Telómero/metabolismo , Ácido Anhídrido Hidrolasas , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Humanos , Proteína Homóloga de MRE11 , Osteosarcoma/metabolismo , Osteosarcoma/ultraestructura , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Células Tumorales Cultivadas , Proteínas de Unión al GTP rap1/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-25960751

RESUMEN

We investigated the effects of Hericium erinaceus (HEM) on liver injury induced by acute alcohol administration in mice. Mice received ethanol (5 g/kg BW) by gavage every 12 hrs for a total of 3 doses. HEM (200 mg/kg BW) was gavage before ethanol administration. Subsequent serum alanine aminotransferase (ALT) level, aspartate aminotransaminase (AST) level, Maleic dialdehyde (MDA) level, hepatic total antioxidant status (TAOS), and activated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) were determined by ELISA and immunohistochemistry, respectively. HEM administration markedly (P < 0.05) decreased serum ALT, AST, and MDA levels. The hepatic histopathological observations showed that HEM had a relatively significant role in mice model, which had alcoholic liver damage. In conclusion, we observed that HEM (200 mg/kg BW) supplementation could restrain the hepatic damage caused by acute alcohol exposure.

11.
PLoS One ; 9(6): e99896, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24959851

RESUMEN

Organization and dynamics of focal adhesion proteins have been well characterized in cells grown on two-dimensional (2D) cell culture surfaces. However, much less is known about the dynamic association of these proteins in the 3D microenvironment. Limited imaging technologies capable of measuring protein interactions in real time and space for cells grown in 3D is a major impediment in understanding how proteins function under different environmental cues. In this study, we applied the nano-scale precise imaging by rapid beam oscillation (nSPIRO) technique and combined the scaning-fluorescence correlation spectroscopy (sFCS) and the number and molecular brightness (N&B) methods to investigate paxillin and actin dynamics at focal adhesions in 3D. Both MDA-MB-231 cells and U2OS cells produce elongated protrusions with high intensity regions of paxillin in cell grown in 3D collagen matrices. Using sFCS we found higher percentage of slow diffusing proteins at these focal spots, suggesting assembling/disassembling processes. In addition, the N&B analysis shows paxillin aggregated predominantly at these focal contacts which are next to collagen fibers. At those sites, actin showed slower apparent diffusion rate, which indicated that actin is either polymerizing or binding to the scaffolds in these locals. Our findings demonstrate that by multiplexing these techniques we have the ability to spatially and temporally quantify focal adhesion assembly and disassembly in 3D space and allow the understanding tumor cell invasion in a more complex relevant environment.


Asunto(s)
Línea Celular/ultraestructura , Adhesiones Focales/química , Adhesiones Focales/metabolismo , Espectrometría de Fluorescencia/métodos , Actinas/metabolismo , Técnicas de Cultivo de Célula , Colágeno Tipo I/química , Humanos , Nanotecnología/métodos , Paxillin/metabolismo , Unión Proteica , Conformación Proteica
12.
EMBO Mol Med ; 5(3): 353-65, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23341130

RESUMEN

RAD51 recombinase activity plays a critical role for cancer cell proliferation and survival, and often contributes to drug-resistance. Abnormally elevated RAD51 function and hyperactive homologous recombination (HR) rates have been found in a panel of cancers, including breast cancer and chronic myeloid leukaemia (CML). Directly targeting RAD51 and attenuating the deregulated RAD51 activity has therefore been proposed as an alternative and supplementary strategy for cancer treatment. Here we show that a newly identified small molecule, IBR2, disrupts RAD51 multimerization, accelerates proteasome-mediated RAD51 protein degradation, reduces ionizing radiation-induced RAD51 foci formation, impairs HR, inhibits cancer cell growth and induces apoptosis. In a murine imatinib-resistant CML model bearing the T315I Bcr-abl mutation, IBR2, but not imatinib, significantly prolonged animal survival. Moreover, IBR2 effectively inhibits the proliferation of CD34(+) progenitor cells from CML patients resistant to known BCR-ABL inhibitors. Therefore, small molecule inhibitors of RAD51 may suggest a novel class of broad-spectrum therapeutics for difficult-to-treat cancers.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Resistencia a Antineoplásicos , Indoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/farmacología , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Recombinasa Rad51/antagonistas & inhibidores , Tetrahidroisoquinolinas/farmacología , Animales , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Femenino , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Humanos , Mesilato de Imatinib , Indoles/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Simulación del Acoplamiento Molecular , Mutación , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Interferencia de ARN , Recombinasa Rad51/química , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Tetrahidroisoquinolinas/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos
13.
Mol Biol Cell ; 22(19): 3584-94, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21832156

RESUMEN

The spindle assemble checkpoint (SAC) is critical for accurate chromosome segregation. Hec1 contributes to chromosome segregation in part by mediating SAC signaling and chromosome alignment. However, the molecular mechanism by which Hec1 modulates checkpoint signaling and alignment remains poorly understood. We found that Hec1 serine 165 (S165) is preferentially phosphorylated at kinetochores. Phosphorylated Hec1 serine 165 (pS165) specifically localized to kinetochores of misaligned chromosomes, showing a spatiotemporal distribution characteristic of SAC molecules. Expressing an RNA interference (RNAi)-resistant S165A mutant in Hec1-depleted cells permitted normal progression to metaphase, but accelerated the metaphase-to-anaphase transition. The S165A cells were defective in Mad1 and Mad2 localization to kinetochores, regardless of attachment status. These cells often entered anaphase with lagging chromosomes and elicited increased segregation errors and cell death. In contrast, expressing S165E mutant in Hec1-depleted cells triggered defective chromosome alignment and severe mitotic arrest associated with increased Mad1/Mad2 signals at prometaphase kinetochores. A small portion of S165E cells eventually bypassed the SAC but showed severe segregation errors. Nek2 is the primary kinase responsible for kinetochore pS165, while PP1 phosphatase may dephosphorylate pS165 during SAC silencing. Taken together, these results suggest that modifications of Hec1 S165 serve as an important mechanism in modulating SAC signaling and chromosome alignment.


Asunto(s)
Segregación Cromosómica , Puntos de Control de la Fase M del Ciclo Celular , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Anafase , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto , Humanos , Cinetocoros/metabolismo , Proteínas Mad2 , Metafase , Mitosis , Datos de Secuencia Molecular , Quinasas Relacionadas con NIMA , Proteínas Nucleares/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/genética
14.
Nat Struct Mol Biol ; 17(9): 1124-35, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20729863

RESUMEN

Class switch DNA recombination (CSR) is the mechanism that diversifies the biological effector functions of antibodies. Activation-induced cytidine deaminase (AID), a key protein in CSR, targets immunoglobulin H (IgH) switch regions, which contain 5'-AGCT-3' repeats in their core. How AID is recruited to switch regions remains unclear. Here we show that 14-3-3 adaptor proteins have an important role in CSR. 14-3-3 proteins specifically bound 5'-AGCT-3' repeats, were upregulated in B cells undergoing CSR and were recruited with AID to the switch regions that are involved in CSR events (Smu-->Sgamma1, Smu-->Sgamma3 or Smu-->Salpha). Moreover, blocking 14-3-3 by difopein, 14-3-3gamma deficiency or expression of a dominant-negative 14-3-3sigma mutant impaired recruitment of AID to switch regions and decreased CSR. Finally, 14-3-3 proteins interacted directly with AID and enhanced AID-mediated in vitro DNA deamination, further emphasizing the important role of these adaptors in CSR.


Asunto(s)
Proteínas 14-3-3/metabolismo , Citidina Desaminasa/metabolismo , Región de Cambio de la Inmunoglobulina , Recombinación Genética , Proteínas 14-3-3/deficiencia , Proteínas 14-3-3/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Línea Celular , Citidina Desaminasa/inmunología , Humanos , Ratones , Unión Proteica
15.
Mol Biol Cell ; 20(22): 4686-95, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19776357

RESUMEN

Previous studies have stipulated Hec1 as a conserved kinetochore component critical for mitotic control in part by directly binding to kinetochore fibers of the mitotic spindle and by recruiting spindle assembly checkpoint proteins Mad1 and Mad2. Hec1 has also been reported to localize to centrosomes, but its function there has yet to be elucidated. Here, we show that Hec1 specifically colocalizes with Hice1, a previously characterized centrosomal microtubule-binding protein, at the spindle pole region during mitosis. In addition, the C-terminal region of Hec1 directly binds to the coiled-coil domain 1 of Hice1. Depletion of Hice1 by small interfering RNA (siRNA) reduced levels of Hec1 in the cell, preferentially at centrosomes and spindle pole vicinity. Reduction of de novo microtubule nucleation from mitotic centrosomes can be observed in cells treated with Hec1 or Hice1 siRNA. Consistently, neutralization of Hec1 or Hice1 by specific antibodies impaired microtubule aster formation from purified mitotic centrosomes in vitro. Last, disruption of the Hec1/Hice1 interaction by overexpressing Hice1DeltaCoil1, a mutant defective in Hec1 interaction, elicited abnormal spindle morphology often detected in Hec1 and Hice1 deficient cells. Together, the results suggest that Hec1, through cooperation with Hice1, contributes to centrosome-directed microtubule growth to facilitate establishing a proper mitotic spindle.


Asunto(s)
Centrosoma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitosis/fisiología , Proteínas Nucleares/metabolismo , Huso Acromático/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Huso Acromático/ultraestructura , Técnicas del Sistema de Dos Híbridos
16.
J Med Chem ; 52(6): 1757-67, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19243176

RESUMEN

High expression in cancer 1 (Hec1) is an oncogene overly expressed in many human cancers. Small molecule inhibitor of Nek2/Hec1 (INH) targeting the Hec1 and its regulator, Nek2, in the mitotic pathway, was identified to inactivate Hec1/Nek2 function mediated by protein degradation that subsequently leads to chromosome mis-segregation and cell death. To further improve the efficacy of INH, a series of INH analogues were designed, synthesized, and evaluated. Among these 33 newly synthesized analogues, three of them, 6, 13, and 21, have 6-8 fold more potent cell killing activity than the previous lead compound INH1. Compounds 6 and 21 were chosen for analyzing the underlying action mechanism. They target directly the Hec1/Nek2 pathway and cause chromosome mis-alignment as well as cell death, a mechanism similar to that of INH1. This initial exploration of structural/functional relationship of INH may advance the progress for developing clinically applicable INH analogue.


Asunto(s)
Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Línea Celular Tumoral , Citometría de Flujo , Humanos , Espectroscopía de Resonancia Magnética , Quinasas Relacionadas con NIMA , Espectrometría de Masa por Ionización de Electrospray
17.
J Biol Chem ; 283(40): 27064-73, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18678873

RESUMEN

In yeast mitochondria, RNA degradation takes place through the coordinated activities of ySuv3 helicase and yDss1 exoribonuclease (mtEXO), whereas in bacteria, RNA is degraded via RNaseE, RhlB, PNPase, and enolase. Yeast lacking the Suv3 component of the mtEXO form petits and undergo a toxic accumulation of omega intron RNAs. Mammalian mitochondria resemble their prokaryotic origins by harboring a polyadenylation-dependent RNA degradation mechanism, but whether SUV3 participates in regulating RNA turnover in mammalian mitochondria is unclear. We found that lack of hSUV3 in mammalian cells subsequently yielded an accumulation of shortened polyadenylated mtRNA species and impaired mitochondrial protein synthesis. This suggests that SUV3 may serve in part as a component of an RNA degradosome, resembling its yeast ancestor. Reduction in the expression levels of oxidative phosphorylation components correlated with an increase in reactive oxygen species generation, whereas membrane potential and ATP production were decreased. These cumulative defects led to pleiotropic effects in mitochondria such as decreased mtDNA copy number and a shift in mitochondrial morphology from tubular to granular, which eventually manifests in cellular senescence or cell death. Thus, our results suggest that SUV3 is essential for maintaining proper mitochondrial function, likely through a conserved role in mitochondrial RNA regulation.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Homeostasis/fisiología , Mitocondrias/metabolismo , Estabilidad del ARN/fisiología , ARN/metabolismo , Adenosina Trifosfato/biosíntesis , Línea Celular Tumoral , Humanos , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/biosíntesis , Fosforilación Oxidativa , Biosíntesis de Proteínas/fisiología , ARN Mitocondrial
18.
Mol Cell Biol ; 28(11): 3652-62, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18362163

RESUMEN

Spindle integrity is critical for efficient mitotic progression and accurate chromosome segregation. Deregulation of spindles often leads to structural and functional aberrations, ultimately promoting segregation errors and aneuploidy, a hallmark of most human cancers. Here we report the characterization of a previously identified human sarcoma antigen (gene located at 19p13.11), Hice1, an evolutionarily nonconserved 46-kDa coiled-coil protein. Hice1 shows distinct cytoplasmic localization and associates with interphase centrosomes and mitotic spindles, preferentially at the spindle pole vicinity. Depletion of Hice1 by RNA interference resulted in abnormal and unstable spindle configurations, mitotic delay at prometaphase and metaphase, and elevated aneuploidy. Conversely, loss of Hice1 had minimal effects on interphase centrosome duplication. We also found that both full-length Hice1 and Hice1-N1, which is composed of 149 amino acids of the N-terminal region, but not the mutant lacking the N-terminal region, exhibited activities of microtubule bundling and stabilization at a near-physiological concentration. Consistently, overexpression of Hice1 rendered microtubule bundles in cells resistant to nocodazole- or cold-treatment-induced depolymerization. These results demonstrate that Hice1 is a novel microtubule-associated protein important for maintaining spindle integrity and chromosomal stability, in part by virtue of its ability to bind, bundle, and stabilize microtubules.


Asunto(s)
Inestabilidad Cromosómica/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/genética , Huso Acromático/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/genética , Datos de Secuencia Molecular , Huso Acromático/ultraestructura
19.
Cancer Res ; 68(20): 8393-9, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18922912

RESUMEN

Hec1 is a conserved mitotic regulator critical for spindle checkpoint control, kinetochore functionality, and cell survival. Overexpression of Hec1 has been detected in a variety of human cancers and is linked to poor prognosis of primary breast cancers. Through a chemical genetic screening, we have identified a small molecule, N-(4-[2,4-dimethyl-phenyl]-thiazol-2-yl)-benzamide (INH1), which specifically disrupts the Hec1/Nek2 interaction via direct Hec1 binding. Treating cells with INH1 triggered reduction of kinetochore-bound Hec1 as well as global Nek2 protein level, consequently leading to metaphase chromosome misalignment, spindle aberrancy, and eventual cell death. INH1 effectively inhibited the proliferation of multiple human breast cancer cell lines in culture (GI(50), 10-21 micromol/L). Furthermore, treatment with INH1 retarded tumor growth in a nude mouse model bearing xenografts derived from the human breast cancer line MDA-MB-468, with no apparent side effects. This study suggests that the Hec1/Nek2 pathway may serve as a novel mitotic target for cancer intervention by small compounds.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Mitosis/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Benzamidas/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas del Citoesqueleto , Femenino , Humanos , Indoles , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Quinasas Relacionadas con NIMA , Trasplante de Neoplasias , Transducción de Señal/efectos de los fármacos , Tiazoles/uso terapéutico , Trasplante Heterólogo , Valeratos
20.
Cell Cycle ; 7(13): 2013-20, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18604178

RESUMEN

Chromokinesins are microtubule-motor molecules that possess chromatin binding activity and are important for mitotic and meiotic regulation. The chromokinesin-member Kif4A is unique in that it localizes to nucleus during interphase of the cell cycle. Kif4 deletion by gene targeting in mouse embryonic cells was known to associate with DNA damage response. However, its precise role in DNA damage or repair pathway is not clear. Here we report that Kif4A associates with BRCA2 in a biochemical identification and that the interaction is mediated by the Kif4A C-terminal cargo-binding domain and BRCA2 C-terminal conserved region. Upon nucleus-specific laser micro-irradiation, Kif4A was rapidly recruited to sites of DNA damage. Significantly, the depletion of Kif4A from cells by shRNA impaired the ionizing-radiation induced foci (IRIF) formation of Rad51, both quantitatively and qualitatively. In contrast, the IRIF of gamma-H2AX or NBS1 was largely intact. Moreover, Kif4A knockdown rendered cells hypersensitive to ionizing radiation in a colonogenic survival assay. We further demonstrated that Kif4A deficiency led to significantly decreased homologous recombination in an I-SceI endonuclease induced in vivo recombination assay. Together, our results suggest a novel role for a chromokinesin family member in the DNA damage response by modulating the BRCA2/Rad51 pathway.


Asunto(s)
Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Histonas/metabolismo , Cinesinas/metabolismo , Proteínas Nucleares/metabolismo , Recombinasa Rad51/metabolismo , Ciclo Celular , Reparación del ADN , Vectores Genéticos , Células HeLa , Humanos , Recombinación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA