Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Commun ; 12(1): 2705, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976187

RESUMEN

Androgen signaling through the androgen receptor (AR) directs gene expression in both normal and prostate cancer cells. Androgen regulates multiple aspects of the AR life cycle, including its localization and post-translational modification, but understanding how modifications are read and integrated with AR activity has been difficult. Here, we show that ADP-ribosylation regulates AR through a nuclear pathway mediated by Parp7. We show that Parp7 mono-ADP-ribosylates agonist-bound AR, and that ADP-ribosyl-cysteines within the N-terminal domain mediate recruitment of the E3 ligase Dtx3L/Parp9. Molecular recognition of ADP-ribosyl-cysteine is provided by tandem macrodomains in Parp9, and Dtx3L/Parp9 modulates expression of a subset of AR-regulated genes. Parp7, ADP-ribosylation of AR, and AR-Dtx3L/Parp9 complex assembly are inhibited by Olaparib, a compound used clinically to inhibit poly-ADP-ribosyltransferases Parp1/2. Our study reveals the components of an androgen signaling axis that uses a writer and reader of ADP-ribosylation to regulate protein-protein interactions and AR activity.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Poli(ADP-Ribosa) Polimerasas/genética , Neoplasias de la Próstata/genética , Procesamiento Proteico-Postraduccional , Receptores Androgénicos/genética , ADP-Ribosilación/efectos de los fármacos , Adenocarcinoma , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Masculino , Metribolona/farmacología , Proteínas de Neoplasias/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Análisis de Supervivencia
2.
iScience ; 23(12): 101855, 2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-33313495

RESUMEN

Anti-androgens are a common therapy in prostate cancer (PCa) targeting androgen receptor (AR) signaling. However, these therapies fail due to selection of highly aggressive AR-negative cancer cells that have no therapeutic options available. We demonstrate that elevating endogenous ceramide levels with administration of exogenous ceramide nanoliposomes (CNLs) was efficacious in AR-negative cell lines with limited efficacy in AR-positive cells. This effect is mediated through reduced de novo sphingolipid synthesis in AR-positive cells. We show that anti-androgens elevate de novo generation of sphingolipids via SPTSSB, a rate-limiting mediator of sphingolipid generation. Moreover, pharmacological inhibition of AR increases the efficacy of CNL in AR-positive cells through de novo synthesis, while SPTSSB knockdown limited CNL's efficacy in AR-negative cells. Alluding to clinical relevance, SPTSSB is upregulated in patients with advanced PCa after anti-androgens treatment. These findings emphasize the relevance of AR regulation upon sphingolipid metabolism and the potential of CNL as a PCa therapeutic.

3.
Nat Biotechnol ; 38(11): 1317-1327, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32541958

RESUMEN

Current methods can illuminate the genome-wide activity of CRISPR-Cas9 nucleases, but are not easily scalable to the throughput needed to fully understand the principles that govern Cas9 specificity. Here we describe 'circularization for high-throughput analysis of nuclease genome-wide effects by sequencing' (CHANGE-seq), a scalable, automatable tagmentation-based method for measuring the genome-wide activity of Cas9 in vitro. We applied CHANGE-seq to 110 single guide RNA targets across 13 therapeutically relevant loci in human primary T cells and identified 201,934 off-target sites, enabling the training of a machine learning model to predict off-target activity. Comparing matched genome-wide off-target, chromatin modification and accessibility, and transcriptional data, we found that cellular off-target activity was two to four times more likely to occur near active promoters, enhancers and transcribed regions. Finally, CHANGE-seq analysis of six targets across eight individual genomes revealed that human single-nucleotide variation had significant effects on activity at ~15.2% of off-target sites analyzed. CHANGE-seq is a simplified, sensitive and scalable approach to understanding the specificity of genome editors.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Epigénesis Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Secuencia de Bases , Línea Celular , Cromatina/genética , Edición Génica , Variación Genética , Genoma Humano , Humanos , Aprendizaje Automático
4.
Methods Mol Biol ; 1966: 107-124, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31041742

RESUMEN

ADP-ribosylation is a posttranslational modification generated by members of the superfamily of ADP-ribosyltransferases, known as the Parp enzymes. Depending on the superfamily member, Parp enzymes can mono- or poly-ADP-ribosylate a protein substrate. Parp superfamily members confer regulation to a variety of biological processes that include cell signaling, DNA repair, transcription, and stress responses. Here, we describe biochemical methods for detection of ADP-ribose conjugated to the androgen receptor (AR) using the archaeal macrodomain, AF1521, from Archaeoglobus fulgidus. The utility of AF1521 is based on its highly selective recognition of ADP-ribose conjugated to protein. AF1521 immobilized on beads can be used to enrich for ADP-ribosylated proteins, which in our application results in recovery of ADP-ribosylated AR from prostate cancer cell extracts. We engineered tandem AF1521 macrodomains and found this improves the recovery of ADP-ribosylated AR under native conditions, and it enabled development of an assay for detection of ADP-ribosylation on blots. Thus, AF1521 can be used to query ADP-ribosylation of protein under both native and denaturing conditions. Our assays should prove useful for understanding how ADP-ribosylation regulates AR function.


Asunto(s)
ADP-Ribosilación , Técnicas In Vitro/métodos , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/análisis , Receptores Androgénicos/metabolismo , Adenosina Difosfato Ribosa/análisis , Proteínas Arqueales , Archaeoglobus fulgidus/metabolismo , Línea Celular Tumoral , Humanos , Masculino
5.
Aging Cell ; 18(1): e12851, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30565836

RESUMEN

The Ran GTPase regulates nuclear import and export by controlling the assembly state of transport complexes. This involves the direct action of RanGTP, which is generated in the nucleus by the chromatin-associated nucleotide exchange factor, RCC1. Ran interactions with RCC1 contribute to formation of a nuclear:cytoplasmic (N:C) Ran protein gradient in interphase cells. In previous work, we showed that the Ran protein gradient is disrupted in fibroblasts from Hutchinson-Gilford progeria syndrome (HGPS) patients. The Ran gradient disruption in these cells is caused by nuclear membrane association of a mutant form of Lamin A, which induces a global reduction in heterochromatin marked with Histone H3K9me3 and Histone H3K27me3. Here, we have tested the hypothesis that heterochromatin controls the Ran gradient. Chemical inhibition and depletion of the histone methyltransferases (HMTs) G9a and GLP in normal human fibroblasts reduced heterochromatin levels and caused disruption of the Ran gradient, comparable to that observed previously in HGPS fibroblasts. HMT inhibition caused a defect in nuclear localization of TPR, a high molecular weight protein that, owing to its large size, displays a Ran-dependent import defect in HGPS. We reasoned that pathways dependent on nuclear import of large proteins might be compromised in HGPS. We found that nuclear import of ATM requires the Ran gradient, and disruption of the Ran gradient in HGPS causes a defect in generating nuclear γ-H2AX in response to ionizing radiation. Our data suggest a lamina-chromatin-Ran axis is important for nuclear transport regulation and contributes to the DNA damage response.


Asunto(s)
Cromatina/metabolismo , Daño del ADN , Lámina Nuclear/metabolismo , Transducción de Señal , Proteína de Unión al GTP ran/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Azepinas/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Histonas/metabolismo , Humanos , Interfase/efectos de los fármacos , Lamina Tipo A/metabolismo , Lisina/metabolismo , Metilación/efectos de los fármacos , Lámina Nuclear/efectos de los fármacos , Progeria/patología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos
6.
Methods ; 157: 66-79, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30419333

RESUMEN

The discovery and validation of protein-protein interactions provides a knowledge base that is critical for defining protein networks and how they underpin the biology of the cell. Identification of protein interactions that are highly transient, or sensitive to biochemical disruption, can be very difficult. This challenge has been met by proximity labeling methods which generate reactive species that chemically modify neighboring proteins. The most widely used proximity labeling method is BioID, which features a mutant biotin ligase BirA(Arg118Gly), termed BirA*, fused to a protein of interest. Here, we explore how amino acid substitutions at Arg118 affect the biochemical properties of BirA. We found that relative to wild-type BirA, the Arg118Lys substitution both slightly reduced biotin affinity and increased the release of reactive biotinyl-5'-AMP. BioID using a BirA(Arg118Lys)-Lamin A fusion enabled identification of PCNA as a lamina-proximal protein in HEK293T cells, a finding that was validated by immunofluorescence microscopy. Our data expand on the concept that proximity labeling by BirA fused to proteins of interest can be modulated by amino acid substitutions that affect biotin affinity and the release of biotinyl-5'-AMP.


Asunto(s)
Biotina/química , Biotinilación/métodos , Ligasas de Carbono-Nitrógeno/química , Proteínas de Escherichia coli/química , Proteínas Represoras/química , Biotina/genética , Ligasas de Carbono-Nitrógeno/genética , Escherichia coli/enzimología , Proteínas de Escherichia coli/genética , Células HEK293 , Humanos , Mapas de Interacción de Proteínas/genética , Proteínas Represoras/genética
7.
BMC Cancer ; 18(1): 960, 2018 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305041

RESUMEN

BACKGROUND: The cellular effects of androgen are transduced through the androgen receptor, which controls the expression of genes that regulate biosynthetic processes, cell growth, and metabolism. Androgen signaling also impacts DNA damage signaling through mechanisms involving gene expression and transcription-associated DNA damaging events. Defining the contributions of androgen signaling to DNA repair is important for understanding androgen receptor function, and it also has translational implications. METHODS: We generated RNA-seq data from multiple prostate cancer lines and used bioinformatic analyses to characterize androgen-regulated gene expression. We compared the results from cell lines with gene expression data from prostate cancer xenografts, and patient samples, to query how androgen signaling and prostate cancer progression influences the expression of DNA repair genes. We performed whole genome sequencing to help characterize the status of the DNA repair machinery in widely used prostate cancer lines. Finally, we tested a DNA repair enzyme inhibitor for effects on androgen-dependent transcription. RESULTS: Our data indicates that androgen signaling regulates a subset of DNA repair genes that are largely specific to the respective model system and disease state. We identified deleterious mutations in the DNA repair genes RAD50 and CHEK2. We found that inhibition of the DNA repair enzyme MRE11 with the small molecule mirin inhibits androgen-dependent transcription and growth of prostate cancer cells. CONCLUSIONS: Our data supports the view that crosstalk between androgen signaling and DNA repair occurs at multiple levels, and that DNA repair enzymes in addition to PARPs, could be actionable targets in prostate cancer.


Asunto(s)
Andrógenos/metabolismo , Reparación del ADN/genética , ADN de Neoplasias/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Células PC-3 , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
Prostate ; 77(15): 1452-1467, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28875501

RESUMEN

BACKGROUND: Phosphoinositide-3 (PI-3) kinase signaling has a pervasive role in cancer. One of the key effectors of PI-3 kinase signaling is AKT, a kinase that promotes growth and survival in a variety of cancers. Genetically engineered mouse models of prostate cancer have shown that AKT signaling is sufficient to induce prostatic epithelial neoplasia (PIN), but insufficient for progression to adenocarcinoma. This contrasts with the phenotype of mice with prostate-specific deletion of Pten, where excessive PI-3 kinase signaling induces both PIN and locally invasive carcinoma. We reasoned that additional PI-3 kinase effector kinases promote prostate cancer progression via activities that provide biological complementarity to AKT. We focused on the PKN kinase family members, which undergo activation in response to PI-3 kinase signaling, show expression changes in prostate cancer, and contribute to cell motility pathways in cancer cells. METHODS: PKN kinase activity was measured by incorporation of 32 P into protein substrates. Phosphorylation of the turn-motif (TM) in PKN proteins by mTOR was analyzed using the TORC2-specific inhibitor torin and a PKN1 phospho-TM-specific antibody. Amino acid substitutions in the TM of PKN were engineered and assayed for effects on kinase activity. Cell motility-related functions and PKN localization was analyzed by depletion approaches and immunofluorescence microscopy, respectively. The contribution of PKN proteins to prostate tumorigenesis was characterized in several mouse models that express PKN transgenes. The requirement for PKN activity in prostate cancer initiated by loss of phosphatase and tensin homolog deleted on chromosome 10 (Pten), and the potential redundancy between PKN isoforms, was analyzed by prostate-specific deletion of Pkn1, Pkn2, and Pten. RESULTS AND CONCLUSIONS: PKN1 and PKN2 contribute to motility pathways in human prostate cancer cells. PKN1 and PKN2 kinase activity is regulated by TORC2-dependent phosphorylation of the TM, which together with published data indicates that PKN proteins receive multiple PI-3 kinase-dependent inputs. Transgenic expression of active AKT and PKN1 is not sufficient for progression beyond PIN. Moreover, Pkn1 is not required for tumorigenesis initiated by loss of Pten. Triple knockout of Pten, Pkn1, and Pkn2 in mouse prostate results in squamous cell carcinoma, an uncommon but therapy-resistant form of prostate cancer.


Asunto(s)
Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteína Quinasa C/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular/fisiología , Progresión de la Enfermedad , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/genética , Proteína Quinasa C/genética , Serina-Treonina Quinasas TOR/genética
9.
Mol Cell ; 66(4): 503-516.e5, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28525742

RESUMEN

ADP-ribosylation of proteins is emerging as an important regulatory mechanism. Depending on the family member, ADP-ribosyltransferases either conjugate a single ADP-ribose to a target or generate ADP-ribose chains. Here we characterize Parp9, a mono-ADP-ribosyltransferase reported to be enzymatically inactive. Parp9 undergoes heterodimerization with Dtx3L, a histone E3 ligase involved in DNA damage repair. We show that the Dtx3L/Parp9 heterodimer mediates NAD+-dependent mono-ADP-ribosylation of ubiquitin, exclusively in the context of ubiquitin processing by E1 and E2 enzymes. Dtx3L/Parp9 ADP-ribosylates the carboxyl group of Ub Gly76. Because Gly76 is normally used for Ub conjugation to substrates, ADP-ribosylation of the Ub carboxyl terminus precludes ubiquitylation. Parp9 ADP-ribosylation activity therefore restrains the E3 function of Dtx3L. Mutation of the NAD+ binding site in Parp9 increases the DNA repair activity of the heterodimer. Moreover, poly(ADP-ribose) binding to the Parp9 macrodomains increases E3 activity. Dtx3L heterodimerization with Parp9 enables NAD+ and poly(ADP-ribose) regulation of E3 activity.


Asunto(s)
Adenosina Difosfato Ribosa/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Línea Celular Tumoral , Reparación del ADN , Células HEK293 , Humanos , Mutación , NAD/metabolismo , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
10.
Genes Chromosomes Cancer ; 53(12): 963-71, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25131334

RESUMEN

A hallmark of many neoplasias is chromosomal rearrangement, an event that commonly results in the fusion of two separate genes. The RNA and protein resulting from these gene fusions often play critical roles in cancer development, maintenance, and progression. Traditionally, these fusion products are thought to be produced solely due to DNA level changes and are therefore considered unique to cancer. Recent advances in microarray and deep-sequencing have revealed many more fusion transcripts. Surprisingly, some are without detectable rearrangement at the DNA level. Reports have demonstrated that at least some of these chimeric RNAs are generated via intergenic splicing. In this review, we highlight three examples of these noncanonical chimeric transcripts that are formed by trans-splicing or cis-splicing of adjacent genes and summarize the knowledge we have regarding these noncanonical fusions. We discuss the implications of the chimeric RNAs in both cancer and normal physiology, as some of these fusion transcripts are found in normal, noncancerous cells with sequences identical to those generated by canonical chromosomal translocation found in cancer cells. Finally, we present methods that are currently being used to discover additional chimeric RNAs.


Asunto(s)
Fusión Génica , Neoplasias/genética , Empalme del ARN , ARN/genética , Aberraciones Cromosómicas , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Masculino , Desarrollo de Músculos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN/metabolismo , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Sarcoma Estromático Endometrial/genética , Sarcoma Estromático Endometrial/metabolismo , Trans-Empalme
11.
J Vis Exp ; (87)2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24894444

RESUMEN

Many efforts have been devoted to establish in vitro cell culture systems. These systems are designed to model a vast number of in vivo processes. Cell culture systems arising from human endometrial samples are no exception. Applications range from normal cyclic physiological processes to endometrial pathologies such as gynecological cancers, infectious diseases, and reproductive deficiencies. Here, we provide two methods for establishing primary endometrial stromal cells from surgically resected endometrial hysterectomy specimens. The first method is referred to as "the scraping method" and incorporates mechanical scraping using surgical or razor blades whereas the second method is termed "the trypsin method." This latter method uses the enzymatic activity of trypsin to promote the separation of cells and primary cell outgrowth. We illustrate step-by-step methodology through digital images and microscopy. We also provide examples for validating endometrial stromal cell lines via quantitative real time polymerase chain reactions (qPCR) and immunofluorescence (IF).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Endometrio/citología , Células del Estroma/citología , Línea Celular , Endometrio/cirugía , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Histerectomía , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Proc Natl Acad Sci U S A ; 109(16): 6130-5, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22474389

RESUMEN

Nuclear translocation of cytosolic CLIC4 is an essential feature of its proapoptotic and prodifferentiation functions. Here we demonstrate that CLIC4 is induced concurrently with inducible nitric oxide synthase (iNOS) and S-nitrosylated in proinflammatory peritoneal macrophages. Chemical inhibition or genetic ablation of iNOS inhibits S-nitrosylation and nuclear translocation of CLIC4. In macrophages, iNOS-induced nuclear CLIC4 coincides with the pro- to anti-inflammatory transition of the cells because IL-1ß and CXCL1 mRNA remain elevated in CLIC4 and iNOS knockout macrophages at late time points, whereas TNFα mRNA is elevated only in the iNOS knockout macrophages. Active IL-1ß remains elevated in CLIC4 knockout macrophages and in macrophages in which CLIC4 nuclear translocation is prevented by the NOS inhibitor l-NAME. Moreover, overexpression of nuclear-targeted CLIC4 down-regulates IL-1ß in stimulated macrophages. In mice, genetically null for CLIC4, the number of phagocytosing macrophages stimulated by LPS is reduced. Thus, iNOS-induced nuclear CLIC4 is an essential part of the macrophage deactivation program.


Asunto(s)
Núcleo Celular/metabolismo , Canales de Cloruro/metabolismo , Macrófagos/metabolismo , Proteínas Mitocondriales/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Línea Celular , Células Cultivadas , Canales de Cloruro/genética , Expresión Génica/efectos de los fármacos , Immunoblotting , Interferón gamma/farmacología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Carcinogenesis ; 33(5): 986-95, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22387366

RESUMEN

Chloride intracellular channel (CLIC) 4 is a member of a redox-regulated, metamorphic multifunctional protein family, first characterized as intracellular chloride channels. Current knowledge indicates that CLICs participate in signaling, cytoskeleton integrity and differentiation functions of multiple tissues. In metabolically stressed skin keratinocytes, cytoplasmic CLIC4 is S-nitrosylated and translocates to the nucleus where it enhances transforming growth factor-ß (TGF-ß) signaling by protecting phospho-Smad 2 and 3 from dephosphorylation. CLIC4 expression is diminished in multiple human epithelial cancers, and the protein is excluded from the nucleus. We now show that CLIC4 expression is reduced in chemically induced mouse skin papillomas, mouse and human squamous carcinomas and squamous cancer cell lines, and the protein is excluded from the nucleus. The extent of reduction in CLIC4 coincides with progression of squamous tumors from benign to malignant. Inhibiting antioxidant defense in tumor cells increases S-nitrosylation and nuclear translocation of CLIC4. Adenoviral-mediated reconstitution of nuclear CLIC4 in squamous cancer cells enhances TGF-ß-dependent transcriptional activity and inhibits growth. Adenoviral targeting of CLIC4 to the nucleus of tumor cells in orthografts inhibits tumor growth, whereas elevation of CLIC4 in transgenic epidermis reduces de novo chemically induced skin tumor formation. In parallel, overexpression of exogenous CLIC4 in squamous tumor orthografts suppresses tumor growth and enhances TGF-ß signaling. These results indicate that CLIC4 suppresses the growth of squamous cancers, that reduced CLIC4 expression and nuclear residence detected in cancer cells is associated with the altered redox state of tumor cells and the absence of detectable nuclear CLIC4 in cancers contributes to TGF-ß resistance and enhances tumor development.


Asunto(s)
Canales de Cloruro/biosíntesis , Proteínas Mitocondriales/biosíntesis , Neoplasias de Células Escamosas/metabolismo , Neoplasias Cutáneas/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Fibroblastos/metabolismo , Humanos , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos SENCAR , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neoplasias de Células Escamosas/genética , Oxidación-Reducción , Papiloma/genética , Papiloma/metabolismo , Transporte de Proteínas , Transducción de Señal , Neoplasias Cutáneas/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
14.
J Biol Chem ; 285(31): 23818-28, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20504765

RESUMEN

Nuclear translocation of chloride intracellular channel protein CLIC4 is essential for its role in Ca(2+)-induced differentiation, stress-induced apoptosis, and modulating TGF-beta signaling in mouse epidermal keratinocytes. However, post-translational modifications on CLIC4 that govern nuclear translocation and thus these activities remain to be elucidated. The structure of CLIC4 is dependent on the redox environment, in vitro, and translocation may depend on reactive oxygen and nitrogen species in the cell. Here we show that NO directly induces nuclear translocation of CLIC4 that is independent of the NO-cGMP pathway. Indeed, CLIC4 is directly modified by NO through S-nitrosylation of a cysteine residue, as measured by the biotin switch assay. NO enhances association of CLIC4 with the nuclear import proteins importin alpha and Ran. This is likely a result of the conformational change induced by S-nitrosylated CLIC4 that leads to unfolding of the protein, as exhibited by CD spectra analysis and trypsinolysis of the modified protein. Cysteine mutants of CLIC4 exhibit altered nitrosylation, nuclear residence, and stability, compared with the wild type protein likely as a consequence of altered tertiary structure. Moreover, tumor necrosis factor alpha-induced nuclear translocation of CLIC4 is dependent on nitric-oxide synthase activity. Inhibition of nitric-oxide synthase activity inhibits tumor necrosis factor alpha-induced nitrosylation and association with importin alpha and Ran and ablates CLIC4 nuclear translocation. These results suggest that S-nitrosylation governs CLIC4 structure, its association with protein partners, and thus its intracellular distribution.


Asunto(s)
Transporte Activo de Núcleo Celular , Canales de Cloruro/química , Proteínas Mitocondriales/química , Nitrógeno/química , Animales , Diferenciación Celular , Canales de Cloruro/metabolismo , Queratinocitos/citología , Ratones , Proteínas Mitocondriales/metabolismo , Mutación , Células 3T3 NIH , Óxido Nítrico Sintasa/metabolismo , Oxidación-Reducción , Factor de Necrosis Tumoral alfa/metabolismo , alfa Carioferinas/metabolismo , Proteína de Unión al GTP ran/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA