Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Genes Dev ; 31(15): 1529-1534, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28877934

RESUMEN

Senescent cell accumulation in aging tissues is linked to age-associated diseases and declining function, prompting efforts to eliminate them. Mass spectrometry analysis revealed that DPP4 (dipeptidyl peptidase 4) was selectively expressed on the surface of senescent, but not proliferating, human diploid fibroblasts. Importantly, the differential presence of DPP4 allowed flow cytometry-mediated isolation of senescent cells using anti-DPP4 antibodies. Moreover, antibody-dependent cell-mediated cytotoxicity (ADCC) assays revealed that the cell surface DPP4 preferentially sensitized senescent, but not dividing, fibroblasts to cytotoxicity by natural killer cells. In sum, the selective expression of DPP4 on the surface of senescent cells enables their preferential elimination.


Asunto(s)
Senescencia Celular/fisiología , Dipeptidil Peptidasa 4/metabolismo , Proteínas de la Membrana/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Citotoxicidad Celular Dependiente de Anticuerpos , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Diploidia , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Células Asesinas Naturales/metabolismo , Subgrupos Linfocitarios/enzimología , Espectrometría de Masas , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo
2.
Nucleic Acids Res ; 50(9): 5111-5128, 2022 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-35524559

RESUMEN

During routine genome duplication, many potential replication origins remain inactive or 'dormant'. Such origin dormancy is achieved, in part, by an interaction with the metabolic sensor SIRT1 deacetylase. We report here that dormant origins are a group of consistent, pre-determined genomic sequences that are distinguished from baseline (i.e. ordinarily active) origins by their preferential association with two phospho-isoforms of the helicase component MCM2. During normal unperturbed cell growth, baseline origins, but not dormant origins, associate with a form of MCM2 that is phosphorylated by DBF4-dependent kinase (DDK) on serine 139 (pS139-MCM2). This association facilitates the initiation of DNA replication from baseline origins. Concomitantly, SIRT1 inhibits Ataxia Telangiectasia and Rad3-related (ATR)-kinase-mediated phosphorylation of MCM2 on serine 108 (pS108-MCM2) by deacetylating the ATR-interacting protein DNA topoisomerase II binding protein 1 (TOPBP1), thereby preventing ATR recruitment to chromatin. In cells devoid of SIRT1 activity, or challenged by replication stress, this inhibition is circumvented, enabling ATR-mediated S108-MCM2 phosphorylation. In turn, pS108-MCM2 enables DDK-mediated phosphorylation on S139-MCM2 and facilitates replication initiation at dormant origins. These observations suggest that replication origin dormancy and activation are regulated by distinct post-translational MCM modifications that reflect a balance between SIRT1 activity and ATR signaling.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Origen de Réplica , Sirtuina 1 , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo
3.
Genes Dev ; 30(10): 1224-39, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27198227

RESUMEN

Some mitochondrial long noncoding RNAs (lncRNAs) are encoded by nuclear DNA, but the mechanisms that mediate their transport to mitochondria are poorly characterized. Using affinity RNA pull-down followed by mass spectrometry analysis, we found two RNA-binding proteins (RBPs), HuR (human antigen R) and GRSF1 (G-rich RNA sequence-binding factor 1), that associated with the nuclear DNA-encoded lncRNA RMRP and mobilized it to mitochondria. In cultured human cells, HuR bound RMRP in the nucleus and mediated its CRM1 (chromosome region maintenance 1)-dependent export to the cytosol. After RMRP was imported into mitochondria, GRSF1 bound RMRP and increased its abundance in the matrix. Loss of GRSF1 lowered the mitochondrial levels of RMRP, in turn suppressing oxygen consumption rates and modestly reducing mitochondrial DNA replication priming. Our findings indicate that RBPs HuR and GRSF1 govern the cytoplasmic and mitochondrial localization of the lncRNA RMRP, which is encoded by nuclear DNA but has key functions in mitochondria.


Asunto(s)
Núcleo Celular/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Mitocondrias/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , ARN Largo no Codificante/metabolismo , Transporte Activo de Núcleo Celular , Células HEK293 , Células HeLa , Humanos , Unión Proteica , Transporte de Proteínas
4.
Nature ; 464(7290): 858-63, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20336070

RESUMEN

Exceptional genomic stability is one of the hallmarks of mouse embryonic stem (ES) cells. However, the genes contributing to this stability remain obscure. We previously identified Zscan4 as a specific marker for two-cell embryo and ES cells. Here we show that Zscan4 is involved in telomere maintenance and long-term genomic stability in ES cells. Only 5% of ES cells express Zscan4 at a given time, but nearly all ES cells activate Zscan4 at least once during nine passages. The transient Zscan4-positive state is associated with rapid telomere extension by telomere recombination and upregulation of meiosis-specific homologous recombination genes, which encode proteins that are colocalized with ZSCAN4 on telomeres. Furthermore, Zscan4 knockdown shortens telomeres, increases karyotype abnormalities and spontaneous sister chromatid exchange, and slows down cell proliferation until reaching crisis by passage eight. Together, our data show a unique mode of genome maintenance in ES cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Inestabilidad Genómica , Telómero/genética , Telómero/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Proliferación Celular , Aberraciones Cromosómicas , Células Madre Embrionarias/citología , Células Madre Embrionarias/patología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Cariotipificación , Meiosis/genética , Meiosis/fisiología , Ratones , Transporte de Proteínas , Recombinación Genética/genética , Intercambio de Cromátides Hermanas/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Regulación hacia Arriba
5.
FASEB J ; 28(1): 382-94, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24081906

RESUMEN

It has been reported that Mitofusin2 (Mfn2) inhibits cell proliferation when overexpressed. We wanted to study the role of endogenous Mfn2 in cell proliferation, along with the structural features of Mfn2 that influence its mitochondrial localization and control of cell proliferation. Mfn2-knockdown clones of a B-cell lymphoma cell line BJAB exhibited an increased rate of cell proliferation. A 2-fold increase in cell proliferation was also observed in Mfn2-knockout mouse embryonic fibroblast (MEF) cells as compared with the control wild-type cells, and the proliferative advantage of the knockout MEF cells was blocked on reintroduction of the Mfn2 gene. Mfn2 exerts its antiproliferative effect by acting as an effector molecule of Ras, resulting in the inhibition of the Ras-Raf-ERK signaling pathway. Furthermore, both the N-terminal (aa 1-264) and the C-terminal (aa 265-757) fragments of Mfn2 blocked cell proliferation through distinct mechanisms: the N-terminal-mediated inhibition was due to its interaction with Raf-1, whereas the C-terminal fragment of Mfn2 inhibited cell proliferation by interacting with Ras. The inhibition of proliferation by the N-terminal fragment was independent of its mitochondrial localization. Collectively, our data provide new insights regarding the role of Mfn2 in controlling cellular proliferation.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas Mitocondriales/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Citometría de Flujo , GTP Fosfohidrolasas/genética , Humanos , Lentivirus/genética , Proteínas Mitocondriales/genética , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología
6.
RNA Biol ; 12(10): 1121-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26325091

RESUMEN

Vascular endothelial growth factor (VEGF) A is a master regulator of neovascularization and angiogenesis. VEGFA is potently induced by hypoxia and by pathological conditions including diabetic retinopathy and tumorigenesis. Fine-tuning of VEGFA expression by different stimuli is important for maintaining tissue vascularization and organ homeostasis. Here, we tested the effect of the hypoxia mimetic cobalt chloride (CoCl2) on VEGFA expression in human cervical carcinoma HeLa cells. We found that CoCl2 increased the levels of VEGFA mRNA and VEGFA protein without affecting VEGFA mRNA stability. Biotin pulldown analysis to capture the RNA-binding proteins (RBPs) bound to VEGFA mRNA followed by mass spectrometry analysis revealed that the RBP HuR [human antigen R, a member of the embryonic lethal abnormal vision (ELAV) family of proteins], interacts with VEGFA mRNA. VEGFA mRNA-tagging experiments showed that exposure to CoCl2 increases the interaction of HuR with VEGFA mRNA and promoted the colocalization of HuR and the distal part of the VEGFA 3'-untranslated region (UTR) in the cytoplasm. We propose that under hypoxia-like conditions, HuR enhances VEGFA mRNA translation.


Asunto(s)
Proteína 1 Similar a ELAV/genética , Biosíntesis de Proteínas , Ribonucleoproteínas/genética , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Hipoxia de la Célula/genética , Cobalto/farmacología , Proteína 1 Similar a ELAV/biosíntesis , Regulación de la Expresión Génica , Células HeLa , Humanos , Espectrometría de Masas , Procesamiento Proteico-Postraduccional , Estabilidad del ARN/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
7.
Aging (Albany NY) ; 16(8): 6717-6730, 2024 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-38637019

RESUMEN

Evaporation of sweat on the skin surface is the major mechanism for dissipating heat in humans. The secretory capacity of sweat glands (SWGs) declines during aging, leading to heat intolerance in the elderly, but the mechanisms responsible for this decline are poorly understood. We investigated the molecular changes accompanying SWG aging in mice, where sweat tests confirmed a significant reduction of active SWGs in old mice relative to young mice. We first identified SWG-enriched mRNAs by comparing the skin transcriptome of Eda mutant Tabby male mice, which lack SWGs, with that of wild-type control mice by RNA-sequencing analysis. This comparison revealed 171 mRNAs enriched in SWGs, including 47 mRNAs encoding 'core secretory' proteins such as transcription factors, ion channels, ion transporters, and trans-synaptic signaling proteins. Among these, 28 SWG-enriched mRNAs showed significantly altered abundance in the aged male footpad skin, and 11 of them, including Foxa1, Best2, Chrm3, and Foxc1 mRNAs, were found in the 'core secretory' category. Consistent with the changes in mRNA expression levels, immunohistology revealed that higher numbers of secretory cells from old SWGs express the transcription factor FOXC1, the protein product of Foxc1 mRNA. In sum, our study identified mRNAs enriched in SWGs, including those that encode core secretory proteins, and altered abundance of these mRNAs and proteins with aging in mouse SWGs.


Asunto(s)
Envejecimiento , Glándulas Sudoríparas , Animales , Glándulas Sudoríparas/metabolismo , Ratones , Envejecimiento/genética , Envejecimiento/metabolismo , Masculino , ARN Mensajero/metabolismo , ARN Mensajero/genética , Transcriptoma
8.
Mol Pharmacol ; 83(1): 157-66, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23066093

RESUMEN

The orphan nuclear receptor estrogen-related receptor alpha (ERRα) directs the transcription of nuclear genes involved in energy homeostasis control and the regulation of mitochondrial mass and function. A crucial role for controlling ERRα-mediated target gene expression has been ascribed to the biarylpyrazole compound 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide (AM251) through direct binding to and destabilization of ERRα protein. Here, we provide evidence that structurally related AM251 analogs also have negative impacts on ERRα protein levels in a cell-type-dependent manner while having no deleterious actions on ERRγ. We show that these off-target cellular effects of AM251 are mediated by proteasomal degradation of nuclear ERRα. Cell treatment with the nuclear export inhibitor leptomycin B did not prevent AM251-induced destabilization of ERRα protein, whereas proteasome inhibition with MG132 stabilized and maintained its DNA-binding function, indicative of ERRα being a target of nuclear proteasomal complexes. NativePAGE analysis revealed that ERRα formed a ∼220-kDa multiprotein nuclear complex that was devoid of ERRγ and the coregulator peroxisome proliferator-activated receptor γ coactivator-1. AM251 induced SUMO-2,3 incorporation in ERRα in conjunction with increased protein kinase C activity, whose activation by phorbol ester also promoted ERRα protein loss. Down-regulation of ERRα by AM251 or small interfering RNA led to increased mitochondria biogenesis while negatively impacting mitochondrial membrane potential. These results reveal a novel molecular mechanism by which AM251 and related compounds alter mitochondrial physiology through destabilization of ERRα.


Asunto(s)
Mitocondrias/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , Receptores de Estrógenos/metabolismo , Línea Celular , Proteínas de Unión al ADN/metabolismo , Metabolismo Energético , Humanos , Potencial de la Membrana Mitocondrial , Mitocondrias/diagnóstico por imagen , Mitocondrias/metabolismo , Complejos Multiproteicos/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Sumoilación , Ultrasonografía , Receptor Relacionado con Estrógeno ERRalfa
9.
Cells ; 12(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37998365

RESUMEN

In metazoans, the largest sirtuin, SIRT1, is a nuclear protein implicated in epigenetic modifications, circadian signaling, DNA recombination, replication, and repair. Our previous studies have demonstrated that SIRT1 binds replication origins and inhibits replication initiation from a group of potential initiation sites (dormant origins). We studied the effects of aging and SIRT1 activity on replication origin usage and the incidence of transcription-replication collisions (creating R-loop structures) in adult human cells obtained at different time points during chronological aging and in cancer cells. In primary, untransformed cells, SIRT1 activity declined and the prevalence of R-loops rose with chronological aging. Both the reduction in SIRT1 activity and the increased abundance of R-loops were also observed during the passage of primary cells in culture. All cells, regardless of donor age or transformation status, reacted to the short-term, acute chemical inhibition of SIRT1 with the activation of excessive replication initiation events coincident with an increased prevalence of R-loops. However, cancer cells activated dormant replication origins, genome-wide, during long-term proliferation with mutated or depleted SIRT1, whereas, in primary cells, the aging-associated SIRT1-mediated activation of dormant origins was restricted to rDNA loci. These observations suggest that chronological aging and the associated decline in SIRT1 activity relax the regulatory networks that protect cells against excess replication and that the mechanisms protecting from replication-transcription collisions at the rDNA loci manifest as differentially enhanced sensitivities to SIRT1 decline and chronological aging.


Asunto(s)
Estructuras R-Loop , Sirtuina 1 , Humanos , ADN Ribosómico/genética , Sirtuina 1/genética , Sirtuina 1/metabolismo , Replicación del ADN/genética , Envejecimiento/genética
10.
J Thromb Haemost ; 20(9): 2098-2108, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35753044

RESUMEN

BACKGROUND: Von Willebrand factor (VWF) is elevated in sickle cell disease (SCD) and contributes to vaso-occlusion through its thrombogenic properties. VWF is regulated by ADAMTS13, a plasma protease that cleaves VWF into less bioactive multimers. Independent investigations have shown VWF to be elevated in SCD, whereas measurements of ADAMTS13 have been variable. OBJECTIVES: We assessed ADAMTS13 activity using multiple activity assays and measured levels of alternative VWF-cleaving proteases in SCD. METHODS/ PATIENTS: Plasma samples were collected from adult patients with SCD (n = 20) at a single institution when presenting for routine red cell exchange transfusion therapy. ADAMTS13 activity was measured by FRETS-VWF73, Technozym ADAMTS-13 Activity ELISA kit and a full-length VWF digestion reaction. Alternative VWF-cleaving proteases were identified by ELISA. A cell culture model was used to study the impact of SCD stimuli on endothelial ADAMTS13 and alternative VWF-cleaving proteases. RESULTS: ADAMTS13 activity was found to be moderately deficient across the SCD cohort as assessed by activity assays using a VWF A2 domain peptide substrate. However, SCD plasma showed preserved ability to digest full-length VWF, suggesting assay-discrepant results. Neutrophil and endothelial-derived proteases were found to be elevated in SCD plasma. Matrix metalloproteinase 9 specifically showed preferential cleavage of full-length VWF. Upregulation of alternative VWF-cleaving proteases occurred in endothelial cells exposed to SCD stimuli such as heme and hypoxia. CONCLUSIONS: This is the first demonstration of accessory plasma enzymes contributing to the regulation of VWF in a specific disease state and may have implications for assessing the VWF/ADAMTS13 axis in other settings.


Asunto(s)
Anemia de Células Falciformes , Factor de von Willebrand , Proteínas ADAM , Proteína ADAMTS13 , Adulto , Células Endoteliales , Humanos , Factor de von Willebrand/química
11.
J Alzheimers Dis ; 87(3): 1251-1290, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35466940

RESUMEN

BACKGROUND: Sporadic Alzheimer's disease (sAD) lacks a unifying hypothesis that can account for the lipid peroxidation observed early in the disease, enrichment of ApoE in the core of neuritic plaques, hallmark plaques and tangles, and selective vulnerability of entorhinal-hippocampal structures. OBJECTIVE: We hypothesized that 1) high expression of ApoER2 (receptor for ApoE and Reelin) helps explain this anatomical vulnerability; 2) lipid peroxidation of ApoE and ApoER2 contributes to sAD pathogenesis, by disrupting neuronal ApoE delivery and Reelin-ApoER2-Dab1 signaling cascades. METHODS: In vitro biochemical experiments; Single-marker and multiplex fluorescence-immunohistochemistry (IHC) in postmortem specimens from 26 individuals who died cognitively normal, with mild cognitive impairment or with sAD. RESULTS: ApoE and ApoER2 peptides and proteins were susceptible to attack by reactive lipid aldehydes, generating lipid-protein adducts and crosslinked ApoE-ApoER2 complexes. Using in situ hybridization alongside IHC, we observed that: 1) ApoER2 is strongly expressed in terminal zones of the entorhinal-hippocampal 'perforant path' projections that underlie memory; 2) ApoE, lipid aldehyde-modified ApoE, Reelin, ApoER2, and the downstream Reelin-ApoER2 cascade components Dab1 and Thr19-phosphorylated PSD95 accumulated in the vicinity of neuritic plaques in perforant path terminal zones in sAD cases; 3) several ApoE/Reelin-ApoER2-Dab1 pathway markers were higher in sAD cases and positively correlated with histological progression and cognitive deficits. CONCLUSION: Results demonstrate derangements in multiple ApoE/Reelin-ApoER2-Dab1 axis components in perforant path terminal zones in sAD and provide proof-of-concept that ApoE and ApoER2 are vulnerable to aldehyde-induced adduction and crosslinking. Findings provide the foundation for a unifying hypothesis implicating lipid peroxidation of ApoE and ApoE receptors in sAD.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteínas E , Moléculas de Adhesión Celular Neuronal , Aldehídos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ligandos , Peroxidación de Lípido , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Placa Amiloide , Receptores de LDL/metabolismo , Proteína Reelina , Serina Endopeptidasas/metabolismo
12.
J Exp Clin Cancer Res ; 41(1): 126, 2022 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-35387667

RESUMEN

BACKGROUND: Compared to normal cells, cancer cells exhibit a higher level of oxidative stress, which primes key cellular and metabolic pathways and thereby increases their resilience under oxidative stress. This higher level of oxidative stress also can be exploited to kill tumor cells while leaving normal cells intact. In this study we have found that isovalerylspiramycin I (ISP I), a novel macrolide antibiotic, suppresses cancer cell growth and tumor metastases by targeting the nucleolar protein selenoprotein H (SELH), which plays critical roles in keeping redox homeostasis and genome stability in cancer cells. METHODS: We developed ISP I through genetic recombination and tested the antitumor effects using primary and metastatic cancer models. The drug target was identified using the drug affinity responsive target stability (DARTS) and mass spectrum assays. The effects of ISP I were assessed for reactive oxygen species (ROS) generation, DNA damage, R-loop formation and its impact on the JNK2/TIF-IA/RNA polymerase I (POLI) transcription pathway. RESULTS: ISP I suppresses cancer cell growth and tumor metastases by targeting SELH. Suppression of SELH induces accumulation of ROS and cancer cell-specific genomic instability. The accumulation of ROS in the nucleolus triggers nucleolar stress and blocks ribosomal RNA transcription via the JNK2/TIF-IA/POLI pathway, causing cell cycle arrest and apoptosis in cancer cells. CONCLUSIONS: We demonstrated that ISP I links cancer cell vulnerability to oxidative stress and RNA biogenesis by targeting SELH. This suggests a potential new cancer treatment paradigm, in which the primary therapeutic agent has minimal side-effects and hence may be useful for long-term cancer chemoprevention.


Asunto(s)
Nucléolo Celular , ARN Ribosómico , Nucléolo Celular/metabolismo , Inestabilidad Genómica , Humanos , Proteínas Nucleares/metabolismo , ARN Ribosómico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo
13.
J Biol Chem ; 285(12): 8887-93, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20103595

RESUMEN

The predominantly nuclear heterogenous ribonucleoprotein A18 (hnRNP A18) translocates to the cytosol in response to cellular stress and increases translation by specifically binding to the 3'-untranslated region (UTR) of several mRNA transcripts and the eukaryotic initiation factor 4G. Here, we identified a 51-nucleotide motif that is present 11.49 times more often in the 3'-UTR of hnRNP A18 mRNA targets than in the UniGene data base. This motif was identified by computational analysis of primary sequences and secondary structures of hnRNP A18 mRNA targets against the unaligned sequences. Band shift analyses indicate that the motif is sufficient to confer binding to hnRNP A18. A search of the entire UniGene data base indicates that the hnRNP A18 motif is also present in the 3'-UTR of the ataxia telangiectasia mutated and Rad3-related (ATR) mRNA. Validation of the predicted hnRNP A18 motif is provided by amplification of endogenous ATR transcript on polysomal fractions immunoprecipitated with hnRNP A18. Moreover, overexpression of hnRNP A18 results in increased ATR protein levels and increased phosphorylation of Chk1, a preferred ATR substrate, in response to UV radiation. In addition, our data indicate that inhibition of casein kinase II or GSK3beta significantly reduced hnRNP A18 cytosolic translocation in response to UV radiation. To our knowledge, this constitutes the first demonstration of a post-transcriptional regulatory mechanism for ATR activity. hnRNP A18 could thus become a new target to trigger ATR activity as back-up stress response mechanisms to functionally compensate for absent or defective responders.


Asunto(s)
Regiones no Traducidas 3' , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas Serina-Treonina Quinasas/genética , ARN/genética , Proteínas Supresoras de Tumor/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Microscopía Confocal/métodos , Datos de Secuencia Molecular , Fosforilación , Homología de Secuencia de Aminoácido
14.
J Biol Chem ; 284(42): 28704-12, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19696445

RESUMEN

Heparan sulfate proteoglycans (HSPGs) are important modulators for optimizing signal transduction of many pathways, including the Wnt pathways. We demonstrate that HSPG glycosaminoglycan levels increased with increasing metastatic potential of melanoma cells. Previous studies have demonstrated that Wnt5A increases the invasiveness of melanoma cells. We further demonstrate that HSPGs potentiate Wnt5A signaling, since enzymatic removal of the HSPG backbone resulted in a decrease in cellular Wnt5A levels, an increase in secreted Wnt5A in cell media, a decrease in downstream signaling, and ultimately, a decrease in invasiveness. Specifically, syndecan 1 and syndecan 4 expression correlated to Wnt5A expression and melanoma malignancy. Knockdown of syndecan 1 or 4 caused decreases in cell invasion, which could be restored by treating the cells with recombinant Wnt5A. These data indicate that syndecan 1 and 4 correlate to increased metastatic potential in melanoma patients and are an important component of the Wnt5A autocrine signaling loop, the activation of which leads to increased metastasis of melanoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteoglicanos de Heparán Sulfato/metabolismo , Melanoma/patología , Proteínas Proto-Oncogénicas/química , Neoplasias Cutáneas/patología , Proteínas Wnt/química , Línea Celular Tumoral , Membrana Celular/metabolismo , Proteoglicanos de Heparán Sulfato/química , Humanos , Melanoma/metabolismo , Metástasis de la Neoplasia , Proteínas Recombinantes/química , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Sindecano-1/metabolismo , Sindecano-4/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt-5a , Cicatrización de Heridas
15.
Nat Commun ; 11(1): 24, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31911655

RESUMEN

The spindle assembly checkpoint (SAC) prevents premature chromosome segregation by inactivating the anaphase promoting complex/cyclosome (APC/C) until all chromosomes are properly attached to mitotic spindles. Here we identify a role for Cullin-RING ubiquitin ligase complex 4 (CRL4), known for modulating DNA replication, as a crucial mitotic regulator that triggers the termination of the SAC and enables chromosome segregation. CRL4 is recruited to chromatin by the replication origin binding protein RepID/DCAF14/PHIP. During mitosis, CRL4 dissociates from RepID and replaces it with RB Binding Protein 7 (RBBP7), which ubiquitinates the SAC mediator BUB3 to enable mitotic exit. During interphase, BUB3 is protected from CRL4-mediated degradation by associating with promyelocytic leukemia (PML) nuclear bodies, ensuring its availability upon mitotic onset. Deficiencies in RepID, CRL4 or RBBP7 delay mitotic exit, increase genomic instability and enhance sensitivity to paclitaxel, a microtubule stabilizer and anti-tumor drug.


Asunto(s)
Anafase , Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metafase , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mitosis , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/metabolismo , Unión Proteica , Proteolisis , Proteína 7 de Unión a Retinoblastoma/genética , Proteína 7 de Unión a Retinoblastoma/metabolismo , Huso Acromático/metabolismo , Ubiquitina-Proteína Ligasas/genética
16.
Brain Behav Immun ; 23(7): 1028-37, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19520156

RESUMEN

Regulatory T cells (Tregs) and beta-galactoside-binding protein (betaGBP), a regulatory protein often found expressed at sites of immunological privilege, have similar functions. Their presence affects the outcome of harmful autoimmunity and cancers, including experimental autoimmune encephalomyelitis and malignant gliomas. Here we report a novel pathway by which Tregs express and utilize betaGBP to control CD8(+) T cell responses partially activating TCR signaling but blocking PI3K activity. As a result, this leads to a loss of p21(ras), ERK and Akt activities despite activation of TCR proximal signals, such as phosphorylation of CD3zeta, Zap70, Lat and PKCtheta. Although non-processive TCR signaling often leads to cell anergy, Tregs/betaGBP did not affect cell viability. Instead, betaGBP/Tregs transiently prevented activation of CD8(+) T cells with self-antigens, while keeping their responses to xenogeneic antigens unaffected.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Galectina 1/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Autoantígenos/inmunología , Autoantígenos/metabolismo , Western Blotting , Linfocitos T CD8-positivos/inmunología , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Citometría de Flujo , Galectina 1/inmunología , Humanos , Microscopía Confocal , Fosforilación/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Proteínas ras/inmunología , Proteínas ras/metabolismo
17.
Int J Med Sci ; 6(2): 93-101, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19305641

RESUMEN

Cytoplasmic expression of claudin-1 in metastatic melanoma cells correlates to increased migration, and increased secretion of MMP-2 in a PKC dependent manner, whereas claudin-1 nuclear expression is found in benign nevi. Melanoma cells were transfected with a vector expressing CLDN-1 fused to a nuclear localization signal (NLS). Despite significant nuclear localization of claudin-1, there was still transport of claudin-1 to the cytoplasm. Phorbol ester treatment of cells transfected with NLS-claudin-1 resulted in an exclusion of claudin-1 from the nucleus, despite the NLS. To ascertain whether PKC or PKA were involved in this translocation, we mutated the putative phosphorylation sites within the protein. We found that mutating the PKC phosphorylation sites to mimic a non-phosphorylated state did not cause a shift of claudin-1 to the nucleus of the cells, but mutating the PKA sites did. Mutations of either site to mimic constitutive phosphorylation resulted in cytoplasmic claudin-1 expression. Stable claudin-1 transfectants containing non-phosphorylatable PKA sites exhibited decreased motility. These data imply that subcellular localization of claudin-1 can be controlled by phosphorylation, dicating effects on metastatic capacity.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Melanoma/patología , Proteínas de la Membrana/metabolismo , Proteína Quinasa C/metabolismo , Transporte Biológico/genética , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Claudina-1 , Simulación por Computador , Citoplasma/genética , Citoplasma/metabolismo , Activación Enzimática , Vectores Genéticos , Humanos , Inmunohistoquímica , Metaloproteinasa 2 de la Matriz/metabolismo , Melanoma/metabolismo , Mutagénesis Sitio-Dirigida , Invasividad Neoplásica , Metástasis de la Neoplasia , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fosforilación , Fracciones Subcelulares/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transfección
18.
Nucleic Acids Res ; 35(15): 4941-51, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17626041

RESUMEN

The Cockayne Syndrome group B (CSB) protein plays important roles in transcription, transcription-coupled nucleotide excision repair and base excision DNA repair. c-Abl kinase also plays a role in DNA repair as a regulator/coordinator of the DNA damage response. This study presents evidence that the N-terminal region of CSB interacts with the SH3 domain of c-Abl in vitro and in vivo. In addition, c-Abl kinase phosphorylates CSB at Tyr932. The subcellular localization of CSB to the nucleus and nucleolus is altered after phosphorylation by c-Abl. c-Abl-dependent phosphorylation of CSB increased in cells treated with hydrogen peroxide and decreased in cells pre-treated with STI-571, a c-Abl-specific protein kinase inhibitor. Activation of the c-Abl kinase in response to oxidative damage is not observed in CSB null cells. These results suggest that c-Abl and CSB may regulate each other in a reciprocal manner in response to oxidative stress.


Asunto(s)
ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Animales , Línea Celular , Células Cultivadas , ADN Helicasas/análisis , ADN Helicasas/química , Enzimas Reparadoras del ADN/análisis , Enzimas Reparadoras del ADN/química , Humanos , Ratones , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Proto-Oncogénicas c-abl/análisis , Tirosina/metabolismo
19.
Mol Cell Biol ; 25(17): 7625-36, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16107709

RESUMEN

Cockayne syndrome (CS) is a rare genetic disorder characterized as a segmental premature-aging syndrome. The CS group B (CSB) protein has previously been implicated in transcription-coupled repair, transcriptional elongation, and restoration of RNA synthesis after DNA damage. Recently, evidence for a role of CSB in base excision repair of oxidative DNA lesions has accumulated. In our search to understand the molecular function of CSB in this process, we identify a physical and functional interaction between CSB and poly(ADP-ribose) polymerase-1 (PARP-1). PARP-1 is a nuclear enzyme that protects the integrity of the genome by responding to oxidative DNA damage and facilitating DNA repair. PARP-1 binds to single-strand DNA breaks which activate the catalytic ability of PARP-1 to add polymers of ADP-ribose to various proteins. We find that CSB is present at sites of activated PARP-1 after oxidative stress, identify CSB as a new substrate of PARP-1, and demonstrate that poly(ADP-ribosyl)ation of CSB inhibits its DNA-dependent ATPase activity. Furthermore, we find that CSB-deficient cell lines are hypersensitive to inhibition of PARP. Our results implicate CSB in the PARP-1 poly(ADP-ribosyl)ation response after oxidative stress and thus suggest a novel role of CSB in the cellular response to oxidative damage.


Asunto(s)
Síndrome de Cockayne , ADN Helicasas/metabolismo , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/genética , Enzimas Reparadoras del ADN , Humanos , Mutación/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Especificidad por Sustrato
20.
Mol Cell Biol ; 25(21): 9520-31, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16227602

RESUMEN

The RNA-binding protein TIA-1 (T-cell intracellular antigen 1) functions as a posttranscriptional regulator of gene expression and aggregates to form stress granules following cellular damage. TIA-1 was previously shown to bind mRNAs encoding tumor necrosis factor alpha (TNF-alpha) and cyclooxygenase 2 (COX-2), but TIA-1 target mRNAs have not been systematically identified. Here, immunoprecipitation (IP) of TIA-1-RNA complexes, followed by microarray-based identification and computational analysis of bound transcripts, was used to elucidate a common motif present among TIA-1 target mRNAs. The predicted TIA-1 motif was a U-rich, 30- to 37-nucleotide (nt)-long bipartite element forming loops of variable size and a bent stem. The TIA-1 motif was found in the TNF-alpha and COX-2 mRNAs and in 3,019 additional UniGene transcripts (approximately 3% of the UniGene database), localizing preferentially to the 3' untranslated region. The interactions between TIA-1 and target transcripts were validated by IP of endogenous mRNAs, followed by reverse transcription and PCR-mediated detection, and by pulldown of biotinylated RNAs, followed by Western blotting. Further studies using RNA interference revealed that TIA-1 repressed the translation of bound mRNAs. In summary, we report a signature motif present in mRNAs that associate with TIA-1 and provide support to the notion that TIA-1 represses the translation of target transcripts.


Asunto(s)
ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Secuencia de Bases , Línea Celular Tumoral , Biología Computacional , Ciclooxigenasa 2/genética , Humanos , Inmunoprecipitación , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA