Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Oncogene ; 41(44): 4841-4854, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36171271

RESUMEN

Pharmacologic inhibition of LSD1 induces molecular and morphologic differentiation of blast cells in acute myeloid leukemia (AML) patients harboring MLL gene translocations. In addition to its demethylase activity, LSD1 has a critical scaffolding function at genomic sites occupied by the SNAG domain transcription repressor GFI1. Importantly, inhibitors block both enzymatic and scaffolding activities, in the latter case by disrupting the protein:protein interaction of GFI1 with LSD1. To explore the wider consequences of LSD1 inhibition on the LSD1 protein complex we applied mass spectrometry technologies. We discovered that the interaction of the HMG-box protein HMG20B with LSD1 was also disrupted by LSD1 inhibition. Downstream investigations revealed that HMG20B is co-located on chromatin with GFI1 and LSD1 genome-wide; the strongest HMG20B binding co-locates with the strongest GFI1 and LSD1 binding. Functional assays demonstrated that HMG20B depletion induces leukemia cell differentiation and further revealed that HMG20B is required for the transcription repressor activity of GFI1 through stabilizing LSD1 on chromatin at GFI1 binding sites. Interaction of HMG20B with LSD1 is through its coiled-coil domain. Thus, HMG20B is a critical component of the GFI1:LSD1 transcription repressor complex which contributes to leukemia cell differentiation block.


Asunto(s)
Histona Demetilasas , Leucemia Mieloide Aguda , Humanos , Diferenciación Celular/genética , Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Demetilasas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Anal Chem ; 94(20): 7339-7349, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35549087

RESUMEN

Large RNA including mRNA (mRNA) has emerged as an important new class of therapeutics. Recently, this has been demonstrated by two highly efficacious vaccines based on mRNA sequences encoding for a modified version of the SARS-CoV-2 spike protein. There is currently significant demand for the development of new and improved analytical methods for the characterization of large RNA including mRNA therapeutics. In this study, we have developed an automated, high-throughput workflow for the rapid characterization and direct sequence mapping of large RNA and mRNA therapeutics. Partial RNase digestions using RNase T1 immobilized on magnetic particles were performed in conjunction with high-resolution liquid chromatography-mass spectrometry analysis. Sequence mapping was performed using automated oligoribonucleotide annotation and identifications based on MS/MS spectra. Using this approach, a >80% sequence of coverage of a range of large RNAs and mRNA therapeutics including the SARS-CoV-2 spike protein was obtained in a single analysis. The analytical workflow, including automated sample preparation, can be completed within 90 min. The ability to rapidly identify, characterize, and sequence map large mRNA therapeutics with high sequence coverage provides important information for identity testing, sequence validation, and impurity analysis.


Asunto(s)
COVID-19 , Espectrometría de Masas en Tándem , COVID-19/terapia , Humanos , ARN/química , ARN Mensajero/genética , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus , Espectrometría de Masas en Tándem/métodos
3.
Nat Commun ; 12(1): 4859, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381042

RESUMEN

Stem and progenitor cells undergo a global elevation of nascent transcription, or hypertranscription, during key developmental transitions involving rapid cell proliferation. The chromatin remodeler Chd1 mediates hypertranscription in pluripotent cells but its mechanism of action remains poorly understood. Here we report a novel role for Chd1 in protecting genome integrity at promoter regions by preventing DNA double-stranded break (DSB) accumulation in ES cells. Chd1 interacts with several DNA repair factors including Atm, Parp1, Kap1 and Topoisomerase 2ß and its absence leads to an accumulation of DSBs at Chd1-bound Pol II-transcribed genes and rDNA. Genes prone to DNA breaks in Chd1 KO ES cells are longer genes with GC-rich promoters, a more labile nucleosomal structure and roles in chromatin regulation, transcription and signaling. These results reveal a vulnerability of hypertranscribing stem cells to accumulation of endogenous DNA breaks, with important implications for developmental and cancer biology.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética , Animales , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Ribosómico/metabolismo , Proteínas de Unión al ADN/genética , Ratones , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Transducción de Señal , Sitio de Iniciación de la Transcripción
4.
Leukemia ; 34(2): 427-440, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31611628

RESUMEN

Inappropriate localization of proteins can interfere with normal cellular function and drive tumor development. To understand how this contributes to the development of acute myeloid leukemia (AML), we compared the nuclear proteome and transcriptome of AML blasts with normal human CD34+ cells. Analysis of the proteome identified networks and processes that significantly affected transcription regulation including misexpression of 11 transcription factors with seven proteins not previously implicated in AML. Transcriptome analysis identified changes in 40 transcription factors but none of these were predictive of changes at the protein level. The highest differentially expressed protein in AML nuclei compared with normal CD34+ nuclei (not previously implicated in AML) was S100A4. In an extended cohort, we found that over-expression of nuclear S100A4 was highly prevalent in AML (83%; 20/24 AML patients). Knock down of S100A4 in AML cell lines strongly impacted their survival whilst normal hemopoietic stem progenitor cells were unaffected. These data are the first analysis of the nuclear proteome in AML and have identified changes in transcription factor expression or regulation of transcription that would not have been seen at the mRNA level. These data also suggest that S100A4 is essential for AML survival and could be a therapeutic target in AML.


Asunto(s)
Núcleo Celular/genética , Leucemia Mieloide Aguda/genética , Proteoma/genética , Proteína de Unión al Calcio S100A4/genética , Transcriptoma/genética , Adolescente , Adulto , Anciano , Antígenos CD34/genética , Proliferación Celular/genética , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Proteómica/métodos
5.
Nucleic Acids Res ; 46(15): 7662-7674, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29939287

RESUMEN

The transcriptional regulator EVI1 has an essential role in early hematopoiesis and development. However, aberrantly high expression of EVI1 has potent oncogenic properties and confers poor prognosis and chemo-resistance in leukemia and solid tumors. To investigate to what extent EVI1 function might be regulated by post-translational modifications we carried out mass spectrometry- and antibody-based analyses and uncovered an ATM-mediated double phosphorylation of EVI1 at the carboxy-terminal S858/S860 SQS motif. In the presence of genotoxic stress EVI1-WT (SQS), but not site mutated EVI1-AQA was able to maintain transcriptional patterns and transformation potency, while under standard conditions carboxy-terminal mutation had no effect. Maintenance of hematopoietic progenitor cell clonogenic potential was profoundly impaired with EVI1-AQA compared with EVI1-WT, in particular in the presence of genotoxic stress. Exploring mechanistic events underlying these observations, we showed that after genotoxic stress EVI1-WT, but not EVI1-AQA increased its level of association with its functionally essential interaction partner CtBP1, implying a role for ATM in regulating EVI1 protein interactions via phosphorylation. This aspect of EVI1 regulation is therapeutically relevant, as chemotherapy-induced genotoxicity might detrimentally sustain EVI1 function via stress response mediated phosphorylation, and ATM-inhibition might be of specific targeted benefit in EVI1-overexpressing malignancies.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Autorrenovación de las Células/genética , Proteínas de Unión al ADN/genética , Regulación Leucémica de la Expresión Génica , Proteína del Locus del Complejo MDS1 y EV11/genética , Enfermedad Aguda , Oxidorreductasas de Alcohol/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Proteína del Locus del Complejo MDS1 y EV11/química , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Mutación , Fosforilación
6.
Cell Death Dis ; 8(6): e2875, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28617445

RESUMEN

BRCA2 encodes a protein with a fundamental role in homologous recombination that is essential for normal development. Carrier status of mutations in BRCA2 is associated with familial breast and ovarian cancer, while bi-allelic BRCA2 mutations can cause Fanconi anemia (FA), a cancer predisposition syndrome with cellular cross-linker hypersensitivity. Cancers associated with BRCA2 mutations can acquire chemo-resistance on relapse. We modeled acquired cross-linker resistance with an FA-derived BRCA2-mutated acute myeloid leukemia (AML) platform. Associated with acquired cross-linker resistance was the expression of a functional BRCA2 protein variant lacking exon 5 and exon 7 (BRCA2ΔE5+7), implying a role for BRCA2 splicing for acquired chemo-resistance. Integrated network analysis of transcriptomic and proteomic differences for phenotyping of BRCA2 disruption infers impact on transcription and chromatin remodeling in addition to the DNA damage response. The striking overlap with transcriptional profiles of FA patient hematopoiesis and BRCA mutation associated ovarian cancer helps define and explicate the 'BRCAness' profile.


Asunto(s)
Empalme Alternativo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Resistencia a Antineoplásicos , Genes BRCA2 , Mutación , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Daño del ADN , Exones , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Humanos , Intrones , Células K562 , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Recurrencia Local de Neoplasia , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Fenotipo , Empalme del ARN , Transcripción Genética
7.
Oncotarget ; 7(43): 70822-70831, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27683124

RESUMEN

Cancer Biomarkers have the capability to improve patient outcomes. They have potential applications in diagnosis, prognosis, monitoring of disease progression and measuring response to treatment. This type of information is particularly useful in the individualisation of treatment regimens. Biomarkers may take many forms but considerable effort has been made to identify and quantify proteins in biological fluids. However, a major challenge in measuring protein in biological fluids, such as plasma, is the sensitivity of the assay and the complex matrix of proteins present. Furthermore, determining the effect of proteases in disease requires measurement of their activity in biological fluids as quantification of the protein itself may not provide sufficient information. To date little progress has been made towards monitoring activity of proteases in plasma. The protease asparaginyl endopeptidase has been implicated in diseases such as breast cancer, leukaemia and dementia. Here we describe a new approach to sensitively and in a targeted fashion quantify asparaginyl endopeptidase activity in plasma using a synthetic substrate peptide protected from nonspecific hydrolysis using D-amino acids within the structure. Our selected reaction monitoring approach enabled asparaginyl endopeptidase activity to be measured in human plasma with both a high dynamic range and sensitivity. This manuscript describes a paradigm for future development of assays to measure protease activities in biological fluids as biomarkers of disease.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Líquidos Corporales/enzimología , Cisteína Endopeptidasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/enzimología , Adolescente , Biomarcadores de Tumor/sangre , Niño , Preescolar , Cromatografía Liquida , Cisteína Endopeptidasas/sangre , Humanos , Lactante , Espectrometría de Masas/métodos , Péptidos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
8.
Nature ; 534(7607): 341-6, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27281222

RESUMEN

Chronic myeloid leukaemia (CML) arises after transformation of a haemopoietic stem cell (HSC) by the protein-tyrosine kinase BCR-ABL. Direct inhibition of BCR-ABL kinase has revolutionized disease management, but fails to eradicate leukaemic stem cells (LSCs), which maintain CML. LSCs are independent of BCR-ABL for survival, providing a rationale for identifying and targeting kinase-independent pathways. Here we show--using proteomics, transcriptomics and network analyses--that in human LSCs, aberrantly expressed proteins, in both imatinib-responder and non-responder patients, are modulated in concert with p53 (also known as TP53) and c-MYC regulation. Perturbation of both p53 and c-MYC, and not BCR-ABL itself, leads to synergistic cell kill, differentiation, and near elimination of transplantable human LSCs in mice, while sparing normal HSCs. This unbiased systems approach targeting connected nodes exemplifies a novel precision medicine strategy providing evidence that LSCs can be eradicated.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Acetamidas/farmacología , Acetamidas/uso terapéutico , Animales , Antígenos CD34/metabolismo , Azepinas/farmacología , Azepinas/uso terapéutico , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Femenino , Proteínas de Fusión bcr-abl/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Imidazolinas/farmacología , Imidazolinas/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Masculino , Ratones , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/trasplante , Proteómica , Proteínas Proto-Oncogénicas c-myc/deficiencia , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Transcriptoma , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Sci Rep ; 6: 26419, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27226058

RESUMEN

The glucocorticoid receptor (GR), a nuclear receptor and major drug target, has a highly conserved minor splice variant, GRγ, which differs by a single arginine within the DNA binding domain. GRγ, which comprises 10% of all GR transcripts, is constitutively expressed and tightly conserved through mammalian evolution, suggesting an important non-redundant role. However, to date no specific role for GRγ has been reported. We discovered significant differences in subcellular localisation, and nuclear-cytoplasmic shuttling in response to ligand. In addition the GRγ transcriptome and protein interactome was distinct, and with a gene ontology signal for mitochondrial regulation which was confirmed using Seahorse technology. We propose that evolutionary conservation of the single additional arginine in GRγ is driven by a distinct, non-redundant functional profile, including regulation of mitochondrial function.


Asunto(s)
Adenosina Trifosfato/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Receptores de Glucocorticoides/metabolismo , Células A549 , Núcleo Celular/metabolismo , Citoplasma , Evolución Molecular , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Proteómica , Receptores de Glucocorticoides/química
10.
Oncotarget ; 7(10): 10739-55, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26919114

RESUMEN

The thrombopoietin receptor (MPL) has been shown to be mutated (MPL W515L) in myelofibrosis and thrombocytosis yet new approaches to treat this disorder are still required. We have previously shown that transcriptome and proteomic effects do not correlate well in oncogene-mediated leukemogenesis. We therefore investigated the effects of MPL W515L using proteomics. The consequences of MPL W515L expression on over 3300 nuclear and 3500 cytoplasmic proteins were assessed using relative quantification mass spectrometry. We demonstrate that MPL W515L expression markedly modulates the CXCL12/CXCR4/CD45 pathway associated with stem and progenitor cell chemotactic movement. We also demonstrated that MPL W515L expressing cells displayed increased chemokinesis which required the MPL W515L-mediated dysregulation of MYC expression via phosphorylation of the RNA transport protein THOC5 on tyrosine 225. In addition MPL W515L expression induced TGFß secretion which is linked to sphingosine 1-phosphate production and the increased chemokinesis. These studies identify several pathways which offer potential targets for therapeutic intervention in the treatment of MPL W515L-driven malignancy. We validate our approach by showing that CD34+ cells from MPL W515L positive patients display increased chemokinesis and that treatment with a combination of MYC and sphingosine kinase inhibitors leads to the preferential killing of MPL W515L expressing cells.


Asunto(s)
Proteínas Nucleares/metabolismo , Receptores de Trombopoyetina/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Humanos , Ratones , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Proteínas Nucleares/genética , Fosforilación , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Transducción de Señal
11.
Int J Cancer ; 138(12): 2984-92, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26815306

RESUMEN

Ovarian cancer (OC) has the highest mortality of all gynaecological cancers. Early diagnosis offers an approach to achieving better outcomes. We conducted a blinded-evaluation of prospectively collected preclinical serum from participants in the multimodal group of the United Kingdom Collaborative Trial of Ovarian Cancer Screening. Using isobaric tags (iTRAQ) we identified 90 proteins differentially expressed between OC cases and controls. A second targeted mass spectrometry analysis of twenty of these candidates identified Protein Z as a potential early detection biomarker for OC. This was further validated by ELISA analysis in 482 serial serum samples, from 80 individuals, 49 OC cases and 31 controls, spanning up to 7 years prior to diagnosis. Protein Z was significantly down-regulated up to 2 years pre-diagnosis (p = 0.000000411) in 8 of 19 Type I patients whilst in 5 Type II individuals, it was significantly up-regulated up to 4 years before diagnosis (p = 0.01). ROC curve analysis for CA-125 and CA-125 combined with Protein Z showed a statistically significant (p = 0.00033) increase in the AUC from 77 to 81% for Type I and a statistically significant (p= 0.00003) increase in the AUC from 76 to 82% for Type II. Protein Z is a novel independent early detection biomarker for Type I and Type II ovarian cancer; which can discriminate between both types. Protein Z also adds to CA-125 and potentially the Risk of Ovarian Cancer algorithm in the detection of both subtypes.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteínas Sanguíneas/metabolismo , Neoplasias Glandulares y Epiteliales/diagnóstico , Neoplasias Ováricas/diagnóstico , Anciano , Detección Precoz del Cáncer , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Ováricas/sangre , Curva ROC
12.
EBioMedicine ; 2(8): 841-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26425690

RESUMEN

Lung cancer is the most frequent cause of cancer-related death world-wide. Radiotherapy alone or in conjunction with chemotherapy is the standard treatment for locally advanced non-small cell lung cancer (NSCLC). Currently there is no predictive marker with clinical utility to guide treatment decisions in NSCLC patients undergoing radiotherapy. Identification of such markers would allow treatment options to be considered for more effective therapy. To enable the identification of appropriate protein biomarkers, plasma samples were collected from patients with non-small cell lung cancer before and during radiotherapy for longitudinal comparison following a protocol that carries sufficient power for effective discovery proteomics. Plasma samples from patients pre- and during radiotherapy who had survived > 18 mo were compared to the same time points from patients who survived < 14 mo using an 8 channel isobaric tagging tandem mass spectrometry discovery proteomics platform. Over 650 proteins were detected and relatively quantified. Proteins which showed a change during radiotherapy were selected for validation using an orthogonal antibody-based approach. Two of these proteins were verified in a separate patient cohort: values of CRP and LRG1 combined gave a highly significant indication of extended survival post one week of radiotherapy treatment.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteína C-Reactiva/metabolismo , Carcinoma de Pulmón de Células no Pequeñas , Glicoproteínas/sangre , Neoplasias Pulmonares , Proteínas de Neoplasias/sangre , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/radioterapia , Masculino , Tasa de Supervivencia
13.
Proc Natl Acad Sci U S A ; 112(17): 5479-84, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25847991

RESUMEN

The glucocorticoid receptor (GR) is a member of the nuclear receptor superfamily, which controls programs regulating cell proliferation, differentiation, and apoptosis. We have identified an unexpected role for GR in mitosis. We discovered that specifically modified GR species accumulate at the mitotic spindle during mitosis in a distribution that overlaps with Aurora kinases. We found that Aurora A was required to mediate mitosis-driven GR phosphorylation, but not recruitment of GR to the spindle. GR was necessary for mitotic progression, with increased time to complete mitosis, frequency of mitotic aberrations, and death in mitosis observed following GR knockdown. Complementation studies revealed an essential role for the GR ligand-binding domain, but no clear requirement for ligand binding in regulating chromosome segregation. The GR N-terminal domain, and specifically phosphosites S203 and S211, were not required. Reduced GR expression results in a cell cycle phenotype, with isolated cells from mouse and human subjects showing changes in chromosome content over prolonged passage. Furthermore, GR haploinsufficient mice have an increased incidence of tumor formation, and, strikingly, these tumors are further depleted for GR, implying additional GR loss as a consequence of cell transformation. We identified reduced GR expression in a panel of human liver, lung, prostate, colon, and breast cancers. We therefore reveal an unexpected role for the GR in promoting accurate chromosome segregation during mitosis, which is causally linked to tumorigenesis, making GR an authentic tumor suppressor gene.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Segregación Cromosómica , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Humanos , Ratones , Ratones Mutantes , Mitosis/genética , Neoplasias/genética , Neoplasias/patología , Estructura Terciaria de Proteína , Receptores de Glucocorticoides/genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética
14.
Stem Cell Reports ; 4(3): 473-88, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25684225

RESUMEN

Mesenchymal progenitor cells have great therapeutic potential, yet incomplete characterization of their cell-surface interface limits their clinical exploitation. We have employed subcellular fractionation with quantitative discovery proteomics to define the cell-surface interface proteome of human bone marrow mesenchymal stromal/stem cells (MSCs) and human umbilical cord perivascular cells (HUCPVCs). We compared cell-surface-enriched fractions from MSCs and HUCPVCs (three donors each) with adult mesenchymal fibroblasts using eight-channel isobaric-tagging mass spectrometry, yielding relative quantification on >6,000 proteins with high confidence. This approach identified 186 upregulated mesenchymal progenitor biomarkers. Validation of 10 of these markers, including ROR2, EPHA2, and PLXNA2, confirmed upregulated expression in mesenchymal progenitor populations and distinct roles in progenitor cell proliferation, migration, and differentiation. Our approach has delivered a cell-surface proteome repository that now enables improved selection and characterization of human mesenchymal progenitor populations.


Asunto(s)
Antígenos de Superficie/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/metabolismo , Proteoma , Proteómica , Adulto , Biomarcadores , Linaje de la Célula/genética , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Fenotipo , Proteómica/métodos , Interferencia de ARN , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Nicho de Células Madre , Adulto Joven
15.
Nucleic Acids Res ; 42(19): 12249-60, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25274738

RESUMEN

Transcription of immediate early genes (IEGs) in response to extrinsic and intrinsic signals is tightly regulated at multiple stages. It is known that untranslated regions of the RNA can play a role in these processes. Here we show that THOC5, a member of the TREX (transcription/export) complex, plays a role in expression of only a subset of constitutively active genes, however transcriptome analysis reveals that more than 90% of IEG were not induced by serum in THOC5 depleted cells. Furthermore, THOC5 depletion does not influence the expression of the most rapidly induced IEGs, e.g. Fos and Jun. One group of THOC5 target genes, including Id1, Id3 and Wnt11 transcripts, were not released from chromatin in THOC5 depleted cells. Genes in another group, including Myc and Smad7 transcripts, were released with shortening of 3'UTR by alternative cleavage, and were spliced but export was impaired in THOC5 depleted cells. By interactome analysis using THOC5 as bait, we show that upon stimulation with serum THOC5 forms a complex with polyadenylation-specific factor 100 (CPSF100). THOC5 is required for recruitment of CPSF100 to 3'UTR of THOC5 target genes. These data suggest the presence of a novel mechanism for the control of IEG response by THOC5 via 3'end-processing.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Genes Inmediatos-Precoces , Proteínas Nucleares/metabolismo , Procesamiento de Término de ARN 3' , Regiones no Traducidas 3' , Animales , Línea Celular , Ratones , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Empalme del ARN , Proteína smad7/genética , Proteína smad7/metabolismo , Transcripción Genética
16.
PLoS One ; 9(5): e97243, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24836440

RESUMEN

Signal transducers and activators of transcription (STATs) are latent cytoplasmic transcription factors linking extracellular signals to target gene transcription. Hematopoietic cells express two highly conserved STAT5-isoforms (STAT5A/STAT5B), and STAT5 is directly activated by JAK2 downstream of several cytokine receptors and the oncogenic BCR-ABL tyrosine kinase. Using an IL-3-dependent cell line with inducible BCR-ABL-expression we compared STAT5-activation by IL-3 and BCR-ABL in a STAT5-isoform specific manner. RNAi targeting of STAT5B strongly inhibits BCR-ABL-dependent cell proliferation, and STAT5B but not STAT5A is essential for BCL-XL-expression in the presence of BCR-ABL. Although BCR-ABL induces STAT5-tyrosine phosphorylation independent of JAK2-kinase activity, BCR-ABL is less efficient in inducing active STAT5A:STAT5B-heterodimerization than IL-3, leaving constitutive STAT5A and STAT5B-homodimerization unaffected. In comparison to IL-3, nuclear accumulation of a STAT5A-eGFP fusion protein is reduced by BCR-ABL, and BCR-ABL tyrosine kinase activity induces STAT5A-eGFP translocation to the cell membrane and co-localization with the IL-3 receptor. Furthermore, BCR-ABL-dependent phosphorylation of Y682 in STAT5A was detected by mass-spectrometry. Finally, RNAi targeting STAT5B but not STAT5A sensitizes human BCR-ABL-positive cell lines to imatinib-treatment. These data demonstrate differences between IL-3 and BCR-ABL-mediated STAT5-activation and isoform-specific effects, indicating therapeutic options for isoform-specific STAT5-inhibition in BCR-ABL-positive leukemia.


Asunto(s)
Proteínas de Fusión bcr-abl/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Benzamidas , Línea Celular , Proliferación Celular/fisiología , Dimerización , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Humanos , Mesilato de Imatinib , Immunoblotting , Inmunoprecipitación , Interleucina-3/metabolismo , Lentivirus , Espectrometría de Masas , Fosforilación , Piperazinas , Pirimidinas , Interferencia de ARN , Transducción de Señal/genética
17.
J Biol Chem ; 289(13): 8931-46, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24488492

RESUMEN

Glucocorticoids (GC) regulate cell fate and immune function. We identified the metastasis-promoting methyltransferase, metastasis-related methyltransferase 1 (WBSCR22/Merm1) as a novel glucocorticoid receptor (GR) regulator relevant to human disease. Merm1 binds the GR co-activator GRIP1 but not GR. Loss of Merm1 impaired both GR transactivation and transrepression by reducing GR recruitment to its binding sites. This was accompanied by loss of GR-dependent H3K4Me3 at a well characterized promoter. Inflammation promotes GC resistance, in part through the actions of TNFα and IFNγ. These cytokines suppressed Merm1 protein expression by driving ubiquitination of two conserved lysine residues. Restoration of Merm1 expression rescued GR transactivation. Cytokine suppression of Merm1 and of GR function was also seen in human lung explants. In addition, striking loss of Merm1 protein was observed in both inflammatory and neoplastic human lung pathologies. In conclusion, Merm1 is a novel regulator of chromatin structure affecting GR recruitment and function, contributing to loss of GC sensitivity in inflammation, with suppressed expression in pulmonary disease.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Metiltransferasas/metabolismo , Receptores de Glucocorticoides/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Bronquios/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucocorticoides/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Lisina/metabolismo , Metilación/efectos de los fármacos , Metiltransferasas/química , Unión Proteica , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitinación/efectos de los fármacos
18.
Bioinformatics ; 30(4): 549-58, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24344193

RESUMEN

MOTIVATION: Isobaric tag for relative and absolute quantitation (iTRAQ) is a widely used method in quantitative proteomics. A robust data analysis strategy is required to determine protein quantification reliability, i.e. changes due to biological regulation rather than technical variation, so that proteins that are differentially expressed can be identified. METHODS: Samples were created by mixing 5, 10, 15 and 20 µg Escherichia coli cell lysate with 100 µg of cell lysate from mouse, corresponding to expected relative fold changes of one for mouse proteins and from 0.25 to 4 for E.coli proteins. Relative quantification was carried out using eight channel isobaric tagging with iTRAQ reagent, and proteins were identified using a TripleTOF 5600 mass spectrometer. Technical variation inherent in this iTRAQ dataset was systematically investigated. RESULTS: A hierarchical statistical model was developed to use quantitative information at peptide level and protein level simultaneously to estimate variation present in each individual peptide and protein. A novel data analysis strategy for iTRAQ, denoted in short as WHATraq, was subsequently proposed with its performance evaluated by the proportion of E.coli proteins that are successfully identified as differentially expressed. Compared with two benchmark data analysis strategies WHATraq was able to identify at least 62.8% more true positive proteins that are differentially expressed. Further validated using a biological iTRAQ dataset including multiple biological replicates from varied murine cell lines, WHATraq performed consistently and identified 375% more proteins as being differentially expressed among different cell lines than the other data analysis strategies.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Modelos Estadísticos , Péptidos/metabolismo , Proteínas/metabolismo , Proteómica/métodos , Algoritmos , Animales , Células Cultivadas , Proteínas de Escherichia coli/análisis , Ratones , Péptidos/análisis , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Proteínas/análisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Coloración y Etiquetado
19.
Mol Cell Proteomics ; 12(11): 3319-29, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23997015

RESUMEN

CXCL12 governs cellular motility, a process deregulated by hematopoietic stem cell oncogenes such as p210-BCR-ABL. A phosphoproteomics approach to the analysis of a hematopoietic progenitor cell line treated with CXCL12 and the Rac 1 and 2 inhibitor NSC23766 has been employed to objectively discover novel mechanisms for regulation of stem cells in normal and malignant hematopoiesis. The proteomic data sets identified new aspects of CXCL12-mediated signaling and novel features of stem cell regulation. We also identified a novel phosphorylation event in hematopoietic progenitor cells that correlated with motile response and governed by the chemotactic factor CXCL12. The novel phosphorylation site on PTPRC/CD45; a protein tyrosine phosphatase, was validated by raising an antibody to the site and also using a mass spectrometry absolute quantification strategy. Site directed mutagenesis and inhibitor studies demonstrated that this single phosphorylation site governs hematopoietic progenitor cell and lymphoid cell motility, lies downstream from Rac proteins and potentiates Src signaling. We have also demonstrated that PTPRC/CD45 is down-regulated in leukemogenic tyrosine kinase expressing cells. The use of discovery proteomics has enabled further understanding of the regulation of PTPRC/CD45 and its important role in cellular motility in progenitor cells.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Células Madre Hematopoyéticas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Aminoquinolinas/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Antígenos Comunes de Leucocito/química , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteómica , Pirimidinas/farmacología , Transducción de Señal
20.
Cell Stem Cell ; 11(2): 179-94, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22862944

RESUMEN

Many signals must be integrated to maintain self-renewal and pluripotency in embryonic stem cells (ESCs) and to enable induced pluripotent stem cell (iPSC) reprogramming. However, the exact molecular regulatory mechanisms remain elusive. To unravel the essential internal and external signals required for sustaining the ESC state, we conducted a short hairpin (sh) RNA screen of 104 ESC-associated phosphoregulators. Depletion of one such molecule, aurora kinase A (Aurka), resulted in compromised self-renewal and consequent differentiation. By integrating global gene expression and computational analyses, we discovered that loss of Aurka leads to upregulated p53 activity that triggers ESC differentiation. Specifically, Aurka regulates pluripotency through phosphorylation-mediated inhibition of p53-directed ectodermal and mesodermal gene expression. Phosphorylation of p53 not only impairs p53-induced ESC differentiation but also p53-mediated suppression of iPSC reprogramming. Our studies demonstrate an essential role for Aurka-p53 signaling in the regulation of self-renewal, differentiation, and somatic cell reprogramming.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Aurora Quinasa A , Aurora Quinasas , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Madre Embrionarias/citología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Fosforilación , Células Madre Pluripotentes/citología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...