Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
bioRxiv ; 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38585889

RESUMEN

The cellular plasticity of neuroblastoma is defined by a mixture of two major cell states, adrenergic (ADRN) and mesenchymal (MES), which may contribute to therapy resistance. However, how neuroblastoma cells switch cellular states during therapy remains largely unknown and how to eradicate neuroblastoma regardless of their cell states is a clinical challenge. To better understand the lineage switch of neuroblastoma in chemoresistance, we comprehensively defined the transcriptomic and epigenetic map of ADRN and MES types of neuroblastomas using human and murine models treated with indisulam, a selective RBM39 degrader. We showed that cancer cells not only undergo a bidirectional switch between ADRN and MES states, but also acquire additional cellular states, reminiscent of the developmental pliancy of neural crest cells. The lineage alterations are coupled with epigenetic reprogramming and dependency switch of lineage-specific transcription factors, epigenetic modifiers and targetable kinases. Through targeting RNA splicing, indisulam induces an inflammatory tumor microenvironment and enhances anticancer activity of natural killer cells. The combination of indisulam with anti-GD2 immunotherapy results in a durable, complete response in high-risk transgenic neuroblastoma models, providing an innovative, rational therapeutic approach to eradicate tumor cells regardless of their potential to switch cell states.

2.
Genome Biol ; 25(1): 16, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216972

RESUMEN

BACKGROUND: The oncogenic protein HOXA9 plays a critical role in leukemia transformation and maintenance, and its aberrant expression is a hallmark of most aggressive acute leukemia. Although inhibiting the upstream regulators of HOXA9 has been proven as a significant therapeutic intervention, the comprehensive regulation network controlling HOXA9 expression in leukemia has not been systematically investigated. RESULTS: Here, we perform genome-wide CRISPR/Cas9 screening in the HOXA9-driven reporter acute leukemia cells. We identify a poorly characterized RNA-binding protein, RBM5, as the top candidate gene required to maintain leukemia cell fitness. RBM5 is highly overexpressed in acute myeloid leukemia (AML) patients compared to healthy individuals. RBM5 loss triggered by CRISPR knockout and shRNA knockdown significantly impairs leukemia maintenance in vitro and in vivo. Through domain CRISPR screening, we reveal that RBM5 functions through a noncanonical transcriptional regulation circuitry rather than RNA splicing, such an effect depending on DNA-binding domains. By integrative analysis and functional assays, we identify HOXA9 as the downstream target of RBM5. Ectopic expression of HOXA9 rescues impaired leukemia cell proliferation upon RBM5 loss. Importantly, acute protein degradation of RBM5 through auxin-inducible degron system immediately reduces HOXA9 transcription. CONCLUSIONS: We identify RBM5 as a new upstream regulator of HOXA9 and reveal its essential role in controlling the survival of AML. These functional and molecular mechanisms further support RBM5 as a promising therapeutic target for myeloid leukemia treatment.


Asunto(s)
Proteínas de Homeodominio , Leucemia Mieloide Aguda , Humanos , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/metabolismo
3.
Exp Ther Med ; 18(3): 1601-1608, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31410115

RESUMEN

The present study aimed to establish a microRNA (miRNA/miR) signature to predict the prognosis of patients with pancreatic cancer (PC) at the early stage and to investigate the involvement of competing endogenous RNAs (ceRNAs) in PC. Using mature miRNA expression profiles from The Cancer Genome Atlas, differentially expressed miRNAs in tissues derived from patients exhibiting early PC and tissues from healthy individuals were compared. The least absolute shrinkage and selection operator regression method was used to construct a miRNA-based signature for predicting prognosis. The miRNet tool, gene set enrichment analysis (GSEA) and the LncRNADisease database were utilized to explore the mechanistic involvement of ceRNAs. A total of seven downregulated miRNAs in PC (miR-424-5p, miR-139-5p, miR-5586-5p, miR-126-3p, miR-3613-5p, miR-454-3p and miR-1271-5p) were selected to generate a signature. Based on this seven-miRNA signature, it was possible to stratify patients with PC into low- and high-risk groups. The overall survival of the low-risk group was significantly longer than that of the high-risk group (P<0.001). The seven-miRNA signature was able to predict the 2-year-survival rate of patients with early PC with an area under the curve of 0.750. Furthermore, as opposed to routine clinicopathological features, this seven-miRNA signature was an independent prognostic factor according to multivariate Cox regression analysis. GSEA indicated that the extracellular matrix receptor interaction pathway and the transforming growth factor-ß signaling pathway were enriched in the high-risk group. A ceRNA network of the seven-miR signature was constructed. In conclusion, the present study provided a seven-miRNA signature, according to which patients with early PC may be divided into high- and low-risk groups. The ceRNA network of the prognostic signature was preliminarily explored.

4.
Int J Gynecol Cancer ; 24(8): 1366-72, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25188887

RESUMEN

PURPOSE: Because of the rising trend of delayed pregnancies, more and more women remain nulliparous at the diagnosis of breast cancer, and approximately 71% of them desire to conceive after breast cancer treatment. Advances in breast cancer screening have made early diagnosis of breast cancer possible, and many patients have the opportunity to be treated by surgery. In this study, we conducted a meta-analysis to evaluate the effect of pregnancy on patient survival and prognosis after surgical treatment for breast cancer. METHODS: An electronic search was performed in MEDLINE (PubMed), EMBASE, and Web of Science to identify potentially eligible studies published before August 2013. Both fixed-effect and random-effect models were used to calculate the pooled relative risk (PRR). The Q test and I(2) statistics were used to assess the heterogeneity among the studies. RESULTS: A total of 5 studies were included in our meta-analysis. Five hundred fifty-four patients who become pregnant after surgical treatment for breast cancer were compared with a control group of 2354 patients for overall survival (OS). Our analysis demonstrated that pregnancy after surgical treatment for breast cancer had a significant beneficial effect on OS (PRR, 0.78; 95% confidence interval, 0.64-0.95). The disease-free survival outcome also favored patients in the pregnancy group (PRR, 0.87; 95% confidence interval, 0.71-1.08). CONCLUSIONS: This meta-analysis indicates that pregnancy after surgical treatment does not increase the risk of breast cancer recurrence and may actually improve OS.


Asunto(s)
Neoplasias de la Mama/rehabilitación , Neoplasias de la Mama/cirugía , Complicaciones Neoplásicas del Embarazo/etiología , Embarazo/fisiología , Seguridad , Neoplasias de la Mama/epidemiología , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Mastectomía/rehabilitación , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/etiología , Complicaciones Neoplásicas del Embarazo/diagnóstico , Complicaciones Neoplásicas del Embarazo/epidemiología , Pronóstico , Factores de Riesgo , Análisis de Supervivencia
5.
Mol Cancer Res ; 11(7): 689-98, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23594796

RESUMEN

Oncogenic c-Myc plays a critical role in cell proliferation, apoptosis, and tumorigenesis, but the precise mechanisms that drive this activity remain largely unknown. P27Kip1 (CDKN1B) arrests cells in G1, and SAP155 (SF3B1), a subunit of the essential splicing factor 3b (SF3b) subcomplex of the spliceosome, is required for proper P27 pre-mRNA splicing. FUSE-binding protein-interacting repressor (FIR), a splicing variant of PUF60 lacking exon5, is a c-Myc transcriptional target that suppresses the DNA helicase p89 (ERCC3) and is alternatively spliced in colorectal cancer lacking the transcriptional repression domain within exon 2 (FIRΔexon2). FIR and FIRΔexon2 form a homo- or hetero-dimer that complexes with SAP155. Our study indicates that the FIR/FIRΔexon2/SAP155 interaction bridges c-Myc and P27 expression. Knockdown of FIR/FIRΔexon2 or SAP155 reduced p27 expression, inhibited its pre-mRNA splicing, and reduced CDK2/Cyclin E expression. Moreover, spliceostatin A, a natural SF3b inhibitor, markedly inhibited P27 expression by disrupting its pre-mRNA splicing and reduced CDK2/Cyclin E expression. The expression of P89, another FIR target, was increased in excised human colorectal cancer tissues. Knockdown of FIR reduced P89; however, the effects on P27 and P89 expression are not simply or directly related to altered FIR expression levels, indicating that the mechanical or physical interaction of the SAP155/FIR/FIRΔexon2 complex is potentially essential for sustained expression of both P89 and P27. Together, the interaction between SAP155 and FIR/FIRΔexon2 not only integrates cell-cycle progression and c-Myc transcription by modifying P27 and P89 expression but also suggests that the interaction is a potential target for cancer screening and treatment.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Empalme Alternativo/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ciclina E/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Piranos/farmacología , Precursores del ARN/genética , Precursores del ARN/metabolismo , Factores de Empalme de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Compuestos de Espiro/farmacología , Factor de Transcripción TFIIH/metabolismo
6.
EMBO J ; 30(5): 846-58, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21285945

RESUMEN

c-myc and p53 networks control proliferation, differentiation, and apoptosis and are responsive to, and cross-regulate a variety of stresses and metabolic and biosynthetic processes. At c-myc, the far upstream element binding protein (FBP) and FBP-interacting repressor (FIR) program transcription by looping to RNA polymerase II complexes engaged at the promoter. Another FBP partner, JTV1/AIMP2, a structural subunit of a multi-aminoacyl-tRNA synthetase (ARS) complex, has also been reported to stabilize p53 via an apparently independent mechanism. Here, we show that in response to oxidative stress, JTV1 dissociates from the ARS complex, translocates to the nucleus, associates with FBP and co-activates the transcription of a new FBP target, ubiquitin-specific peptidase 29 (USP29). A previously uncharacterized deubiquitinating enzyme, USP29 binds to, cleaves poly-ubiquitin chains from, and stabilizes p53. The accumulated p53 quickly induces apoptosis. Thus, FBP and JTV1 help to coordinate the molecular and cellular response to oxidative stress.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Endopeptidasas/genética , Estrés Oxidativo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , Aminoacil-ARNt Sintetasas/genética , Apoptosis , Biomarcadores/metabolismo , Western Blotting , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Endopeptidasas/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN , Secuencias Reguladoras de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas
7.
Chin Med Sci J ; 25(1): 1-12, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20449947

RESUMEN

OBJECTIVE: To investigate the role of TNF receptor-associated factor 2 (TRAF-2) and TRAF6 in CD40-induced nuclear factor-kappaB (NF-kappaB) signaling pathway and whether CD40 signaling requires TRAF2. METHODS: Human B cell lines were transfected with plasmids expressing wild type TRAF2 or dominant negative TRAF2, TRAF2-shRNA, or TRAF6-shRNA. The activation of NF-kappaB was detected by Western blot, kinase assay, transfactor enzyme-linked immunosorbent assay (ELISA), and fluorescence resonance energy transfer (FRET). Analysis of the role of TRAF-2 and TRAF-6 in CD40-mediated NF-kappaB activity was examined following stimulation with recombinant CD154. RESULTS: TRAF2 induced activity of IkappaB-kinases (IKKalpha, IKKi/epsilon), phosphorylation of IkappaBalpha, as well as nuclear translocation and phosphorylation of p65/RelA. In contrast, TRAF6 strongly induced NF-kappaB activation and nuclear translocation of p65 as well as p50 and c-Rel. Engagement of CD154-induced nuclear translocation of p65 was inhibited by a TRAF6-shRNA, but conversely was enhanced by a TRAF2-shRNA. Examination of direct interactions between CD40 and TRAFs by FRET documented that both TRAF2 and TRAF6 directly interacted with CD40. However, the two TRAFs competed for CD40 binding. CONCLUSIONS: These results indicate that TRAF2 can signal in human B cells, but it is not essential for CD40-mediated NF-kappaB activation. Moreover, TRAF2 can compete with TRAF6 for CD40 binding, and thereby limit the capacity of CD40 engagement to induce NF-kappaB activation.


Asunto(s)
Linfocitos B/fisiología , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Linfocitos B/citología , Antígenos CD40/metabolismo , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Humanos , Quinasa I-kappa B/metabolismo , FN-kappa B/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor 2 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor de Transcripción ReIA/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Mol Cancer Ther ; 5(10): 2474-83, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17041091

RESUMEN

Chemokine receptor CXCR4 (CD184) may play a role in cancer metastasis and is known to form homodimers. However, it is not clear how transmembrane regions (TM) of CXCR4 and receptor homotypic interactions affect the function of CXCR4 in living cells. Using confocal microscopy and flow cytometric analysis, we showed that high levels of CXCR4 are present in the cytoplasm, accompanied by lower expression on the cell surface in CXCR4 transfectants, tumor cells, and normal peripheral blood lymphocytes. CXCR4 homodimers were detected in tumor cells, both on the cell surface membrane and in the cytoplasm using fluorescence resonance energy transfer and photobleaching fluorescence resonance energy transfer to measure energy transfer between CXCR4-CFP and CXCR4-YFP constructs. Disruption of lipid rafts by depletion of cholesterol with methyl-beta-cyclodextrin reduced the interaction between CXCR4 molecules and inhibited malignant cell migration to CXCL12/SDF-1alpha. A synthetic peptide of TM4 of CXCR4 reduced energy transfer between molecules of CXCR4, inhibited CXCL12-induced actin polymerization, and blocked chemotaxis of malignant cells. TM4 also inhibited migration of normal monocytes toward CXCL12. Reduction of CXCR4 energy transfer by the TM4 peptide and methyl-beta-cyclodextrin indicates that interactions between CXCR4s may play important roles in cell migration and suggests that cell surface and intracellular receptor dimers are appropriate targets for control of tumor cell spread. Targeting chemokine receptor oligomerization and signal transduction for the treatment of cancer, HIV-1 infections, and other CXCR4 mediated inflammatory conditions warrants further investigation.


Asunto(s)
Quimiotaxis , Péptidos/farmacología , Receptores CXCR4/metabolismo , Actinas/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Línea Celular Tumoral , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiotaxis/efectos de los fármacos , Colesterol/metabolismo , Citoplasma/metabolismo , Dimerización , Transferencia de Energía , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/genética , Humanos , Ligandos , Proteínas Luminiscentes/genética , Linfocitos/metabolismo , Microdominios de Membrana/metabolismo , Microscopía Confocal , Péptidos/química , Receptores CXCR4/química , Receptores CXCR4/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
9.
Cytometry A ; 69(6): 477-86, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16683263

RESUMEN

BACKGROUND: A number of fluorescent caspase substrates and FRET-based indicators have been developed to study the in vivo activation of caspases, a conserved family of proteases critical in inflammatory, and apoptosis signaling pathways. To date, all substrates have measured only one caspase activity. Here, we describe a FRET-based probe for simultaneously measuring two distinct caspase activities in living cells. METHODS: This probe consists of a CFP-YFP-mRFP fusion protein containing a caspase-3-cleavage motif, DEVD, between CFP and YFP, and a caspase-6-cleavage site, VEID, between YFP and mRFP. DEVDase and VEIDase activities could be assessed simultaneously by monitoring diminished FRET mediated by cleavage of either or both of these protease cleavage sites using flow cytometry. RESULTS: DEVDase and VEIDase activities were completely inhibited by the pan-caspase inhibitor z-VAD-fmk and enhanced by DNA-damaging drugs or by anti-Fas stimulation. DEVD and VEID cleavage specificities were validated by using caspase-3-deficient MCF7-Fas cells and a caspase-6-specific inhibitor. Kinetic analysis with the FRET probe revealed that caspase-3 activation consistently preceded caspase-6 by approximately 30 min following induction of apoptosis. CONCLUSIONS: We have developed a novel FRET-based probe for simultaneous detection of two caspase activities in living cells using flow cytometry. Simultaneous detection of two caspase activities using this probe has clearly provided information of the ordering of caspase-3 and -6 in the apoptotic pathway.


Asunto(s)
Caspasas/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes/química , Apoptosis , Proteínas Bacterianas/genética , Caspasa 3 , Caspasa 6 , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Citometría de Flujo/métodos , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Células Jurkat , Cinética , Proteínas Luminiscentes/genética , Modelos Biológicos , Péptido Hidrolasas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Tumorales Cultivadas , Proteína Fluorescente Roja
10.
J Biol Chem ; 281(16): 11235-49, 2006 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-16436380

RESUMEN

Tumor necrosis factor receptor-associated factor 6 (TRAF6) functions as an adaptor, positively regulating the NF-kappaB pathway. Here we report a new function of human TRAF6, the direct stimulation of apoptosis. The mechanism of apoptosis induction results from the capacity of human TRAF6 to interact and activate caspase 8. Both the C-terminal TRAF domain of human TRAF6, which directly interacts with the death effector domain of pro-caspase 8, and the N-terminal RING domain, which is required for activation of caspase 8, are necessary for the induction of apoptosis. The role of endogenous TRAF6 in regulating apoptosis was confirmed by extinguishing TRAF6 expression with specific small-hairpin RNA that resulted in diminished spontaneous apoptosis and resistance to induced apoptosis. In contrast to the human molecule, murine TRAF6 displayed less ability to induce apoptosis and a greater capacity to stimulate NF-kappaB activity. Human and murine TRAF6 are similar except in the region between zinc finger 5 and the TRAF domains. Reciprocal transfer of this connecting region completely exchanged the ability of human and murine TRAF6 to induce apoptosis and activate NF-kappaB. Unique regions of TRAF6 therefore play an important role in determining cell fate.


Asunto(s)
Apoptosis , Caspasas/metabolismo , FN-kappa B/metabolismo , Factor 6 Asociado a Receptor de TNF/fisiología , Animales , Caspasa 8 , Línea Celular , Linaje de la Célula , Clonación Molecular , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Microscopía Confocal , Microscopía Fluorescente , Plásmidos/metabolismo , Estructura Terciaria de Proteína , ARN/química , Factor 6 Asociado a Receptor de TNF/metabolismo , Factores de Tiempo , Transfección , Dedos de Zinc
11.
Nucleic Acids Res ; 33(6): e61, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15805120

RESUMEN

The availability of protein fluorophores with appropriate spectral properties has made it possible to employ fluorescence resonance energy transfer (FRET) to assess interactions between three proteins microscopically. Flow cytometry offers excellent sensitivity, effective signal separation and the capacity to assess a large number of events, and, therefore, should be an ideal means to explore protein interactions in living cells. Here, we report a flow-cytometric FRET technique that employed both direct energy transfer from CFP-->YFP-->mRFP and donor quenching to assess TRAF2 trimerization in living cells. Initially, a series of fusion proteins incorporating CFP, YFP and mRFP with spacers that did or did not permit FRET were employed to document the magnitude of CFP-->YFP and YFP-->mRFP FRET and to calculate the efficiency of CFP-->YFP-->mRFP two-step FRET. Based upon this, TRAF2 homotrimerization could be detected. This method should have great utility in studying the dynamics of interactions between three specific proteins in vivo.


Asunto(s)
Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes/análisis , Proteínas Luminiscentes/análisis , Factor 2 Asociado a Receptor de TNF/análisis , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Línea Celular , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Proteínas Luminiscentes/genética , Microscopía Confocal , Proteínas Recombinantes de Fusión/análisis , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Proteína Fluorescente Roja
12.
J Biol Chem ; 279(53): 55855-65, 2004 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-15383523

RESUMEN

FRET experiments utilizing confocal microscopy or flow cytometry assessed homo- and heterotrimeric association of human tumor necrosis factor receptor-associated factors (TRAF) in living cells. Following transfection of HeLa cells with plasmids expressing CFP- or YFP-TRAF fusion proteins, constitutive homotypic association of TRAF2, -3, and -5 was observed, as well as heterotypic association of TRAF1-TRAF2 and TRAF3-TRAF5. A novel heterotypic association between TRAF2 and -3 was detected and confirmed by immunoprecipitation in Ramos B cells that constitutively express both TRAF2 and -3. Experiments employing deletion mutants of TRAF2 and TRAF3 revealed that this heterotypic interaction minimally involved the TRAF-C domain of TRAF3 as well as the TRAF-N domain and zinc fingers 4 and 5 of TRAF2. A novel flow cytometric FRET analysis utilizing a two-step approach to achieve linked FRET from CFP to YFP to HcRed established that TRAF2 and -3 constitutively form homo- and heterotrimers. The functional importance of TRAF2-TRAF3 heterotrimerization was demonstrated by the finding that TRAF3 inhibited spontaneous NF-kappaB, but not AP-1, activation induced by TRAF2. Ligation of CD40 on Ramos B cells by recombinant CD154 caused TRAF2 and TRAF3 to dissociate, whereas overexpression of TRAF3 in Ramos B cells inhibited CD154-induced TRAF2-mediated activation of NF-kappaB. Together, these results reveal a novel association between TRAF2 and TRAF3 that is mediated by unique portions of each protein and that specifically regulates activation of NF-kappaB, but not AP-1.


Asunto(s)
FN-kappa B/metabolismo , Factor 2 Asociado a Receptor de TNF/química , Factor 3 Asociado a Receptor de TNF/fisiología , Animales , Linfocitos B/metabolismo , Proteínas Bacterianas/metabolismo , Antígenos CD40/biosíntesis , Antígenos CD40/química , Ligando de CD40/química , ADN Complementario/metabolismo , Dimerización , Activación Enzimática , Citometría de Flujo , Eliminación de Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Mutación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/química , Factor 5 Asociado a Receptor de TNF/metabolismo , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo
13.
Am J Pathol ; 164(6): 1901-13, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15161627

RESUMEN

A molecular probe was developed to monitor caspase activity in living cells by flow cytometry. It consists of CFP and YFP with a peptide linker containing two caspase-cleavage sites (LEVD). Its expression resulted in intense fluorescence resonance energy transfer (FRET), whereas cleavage of this linker by caspases eliminated FRET because of physical separation of the CFP and YFP moieties. Using flow cytometry, cells expressing this probe exhibited two patterns, strong FRET and diminished or absent FRET. The appearance of diminished FRET was inhibited by a pan-caspase inhibitor z-VAD or D->A mutations in the LEVD sequence and was markedly increased by apoptosis-inducing agents, etoposide and camptothecin, or overexpression of a caspase 8-red fluorescent protein fusion protein. Importantly, this probe's ability to monitor caspase activity was comparable with results obtained with fluorogenic substrates or fluorochrome-labeled inhibitors of caspases. Specific caspase inhibitors indicated the probe was highly sensitive to cleavage by caspase 6 and 8, less sensitive to caspase 4, and resistant to other caspases. Activation of caspase 8 by Fas engagement markedly increased the probe's cleavage, whereas treatment of caspase 8-deficient cells with anti-Fas did not increase cleavage. However, staurosporine induced cleavage of the probe in caspase 8-deficient cells by a mechanism that was inhibited by overexpression of bcl-x. Taken together, the data indicate that this caspase-sensitive probe can be used to monitor the basal and apoptosis-related activities of caspases, including an initiator caspase, caspase 8, and effector caspases, such as caspase 6.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Secuencia de Bases , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/farmacología , Monitoreo del Ambiente/métodos , Citometría de Flujo/métodos , Colorantes Fluorescentes , Células HeLa , Humanos , Células Jurkat , Proteínas Luminiscentes/genética , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos Antisentido , Plásmidos
14.
Cytometry A ; 55(2): 71-85, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14505312

RESUMEN

BACKGROUND: Protein interactions at the molecular level can be measured by fluorescence resonance energy transfer (FRET) using a pair of fluorescent proteins, such as CFP and YFP, in which the emission spectrum of CFP significantly overlaps the excitation spectrum of YFP. The resulting energy given off from the donor CFP protein can directly excite the acceptor YFP protein when the proteins are closely approximated. During FRET, there is quenching of the emission of the donor CFP protein that is directly related to the efficiency of energy transfer and inversely proportional to the sixth power of the distance between the donor and acceptor proteins. In this study we describe a new approach to visualize donor CFP quenching during CFP-->YFP FRET and demonstrate how this parameter can be used to calculate FRET efficiency. METHODS: A novel flow cytometric method to detect protein-protein interactions in living cells was developed that utilized assessment of CFP donor quenching during CFP-->YFP FRET by comparing CFP intensity between FRET-positive and -negative populations. To accomplish this, we compared the CFP intensity in FRET-positive and FRET-negative cells within the same population transfected with a CFP/YFP fusion protein, in which the molar ratio of CFP:YFP was one. By using separate lasers to excite CFP and YFP, the detection of FRET was separated from that of YFP. Therefore, after direct excitation, the YFP emission spectrum remained constant in all transfected cells, whereas the emission spectrum of CFP varied with the extent of FRET in individual cells. Specific CFP/YFP fusion constructs were prepared to evaluate this approach. The first one consisted of CFP and YFP separated by two caspase cleavage sites (CFP-LEVD-YFP). A second construct consisted of CFP and YFP separated by a structurally restricted 232-amino acid (aa) spacer. No FRET was observed by transfectants expressing this construct. RESULTS: Transfection of CFP-LEVD-YFP into Hela cells resulted in a FRET-positive population and a FRET-negative one. The appearance of the FRET-negative population was inhibited by the caspase inhibitor z-VAD. Moreover, substituting D for A in the caspase cleavage sites of this probe abolished the FRET-negative population, demonstrating the probe's specificity for caspase activity. Comparison of the CFP emission in the FRET-positive and FRET-negative population was used to document the relationship of FRET to donor quenching and permit the calculation of FRET efficiency and relative molecular distance between CFP and YFP. Similar results were noted when cells transfected with the caspase-sensitive probe (in the presence of z-VAD) were mixed with cells expressing the CFP-YFP construct with the 232-aa spacer and therefore were FRET negative. This demonstrated the validity of calculating CFP donor quenching and FRET efficiency by comparing emission spectra of an unknown construct with that of a known positive control, both expressed by the same population of cells. Using this approach, we confirmed that members of the TNF receptor-associated factor (TRAF) family engaged in both homotypic and heterotypic interactions. CONCLUSIONS: We have established a novel flow cytometric approach to assess donor CFP quenching during CFP-->YFP FRET, which can be used for the calculation of FRET efficiency and relative biological molecular distance between CFP and YFP moieties. This method can be used not only to analyze cells that express a CFP and YFP fusion protein, but also independent CFP-coupled and YFP-coupled interacting proteins.


Asunto(s)
Citometría de Flujo/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Luminiscentes/química , Apoptosis/fisiología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Caspasas/metabolismo , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor 3 Asociado a Receptor de TNF , Factor 5 Asociado a Receptor de TNF
15.
Cytometry A ; 53(1): 39-54, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12701131

RESUMEN

BACKGROUND: Use of distinct green fluorescent protein (GFP) variants permits the study of protein-protein interactions and colocalization in viable transfected cells by fluorescence (Förster) resonance energy transfer (FRET). Flow cytometry is a sensitive method to detect FRET. However, the typical dual-laser methods used in flow cytometric FRET assays are not generally applicable because they require a specialized krypton ultraviolet (UV) laser. The purpose of this work was to develop a flow cytometric method to detect FRET between cyan fluorescent protein (CFP; donor) and yellow fluorescent protein (YFP; acceptor) by using the 458-nm excitation from a single tunable argon-ion laser. METHODS: FUSE-binding protein (FBP) interacting repressor (FIR) and FBP are c-myc transcription factors and are known to interact physically. To examine their interaction within viable cells, FIR and the binding motif of FBP, the FBP central domain (FBPcd), were fused with CFP and YFP, respectively, and this pair of fluorescently-tagged proteins was used to detect FRET in vivo. Cells transfected with expression plasmids encoding a CFP-FIR fusion protein and YFP as a negative control, a CFP-YFP fusion protein as a positive control, or CFP-FIR and YFP-FBPcd fusion proteins were examined for FRET after excitation with a 458-nm line from a tunable argon-ion laser. FRET was measured as the ratio of YFP:CFP emission or as YFP emission at 564-606 nm. Conventional FRET using the 413-nm UV line from a krypton laser was examined for comparison. Fluorescence signals were separated with a customized optical filter configuration using 530-nm shortpass, 500-nm longpass, and 560-nm shortpass dichroics in addition to 488/30 nm (CFP), 530/30 nm (YFP), and 585/42 nm (FRET) bandpass filters. Further, a laser-scanning confocal microscopic photobleach technique was used to document that FRET occurred by showing that the intensity of donor CFP fluorescence increased after its acceptor YFP was photobleached. Steady-state spectrofluorometry was used to confirm and validate the results detected by flow cytometry. RESULTS: Upon excitation with the 458-nm line of the argon-ion laser, the enhancement of the acceptor YFP signal and the decrease of the CFP signal were easily detected in cells transfected with the CFP-YFP construct or CFP-FIR and YFP-FBPcd. Similarly, FRET was detected under these conditions when the YFP emission was assessed at 564-606 nm. A strong correlation was observed between the increase in the YFP:CFP ratio and the YFP emission detected at 564-606 nm, consistent with the conclusion that FRET was detected comparably by both methods. A conventional flow cytometric krypton UV-laser technique was also used to confirm that FRET occurred with the CFP-YFP fusion protein and from CFP-FIR --> YFP-FBPcd. FRET also was confirmed by a confocal photobleaching technique, in which donor CFP intensity was enhanced after its acceptor YFP was photobleached. The flow cytometric and confocal microscopic results were confirmed by spectrofluorometry. CONCLUSION: These results demonstrated the feasibility of flow cytometric detection of FRET signals from CFP to YFP by excitation with the 458-nm line from the tunable argon-ion laser. The method was as efficient as excitation with the krypton UV laser and therefore should make FRET a more generally available flow cytometric technique.


Asunto(s)
Argón/química , Proteínas Bacterianas/química , Citometría de Flujo/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Luminiscentes/química , Western Blotting , Línea Celular , ADN Complementario/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Rayos Láser , Microscopía Confocal/métodos , Modelos Teóricos , Plásmidos/metabolismo , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...