Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
EMBO J ; 42(11): e110902, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37039106

RESUMEN

Oncogenic RAS signaling reprograms gene expression through both transcriptional and post-transcriptional mechanisms. While transcriptional regulation downstream of RAS is relatively well characterized, how RAS post-transcriptionally modulates gene expression to promote malignancy remains largely unclear. Using quantitative RNA interactome capture analysis, we here reveal that oncogenic RAS signaling reshapes the RNA-bound proteomic landscape of pancreatic cancer cells, with a network of nuclear proteins centered around nucleolin displaying enhanced RNA-binding activity. We show that nucleolin is phosphorylated downstream of RAS, which increases its binding to pre-ribosomal RNA (rRNA), boosts rRNA production, and promotes ribosome biogenesis. This nucleolin-dependent enhancement of ribosome biogenesis is crucial for RAS-induced pancreatic cancer cell proliferation and can be targeted therapeutically to inhibit tumor growth. Our results reveal that oncogenic RAS signaling drives ribosome biogenesis by regulating the RNA-binding activity of nucleolin and highlight a crucial role for this mechanism in RAS-mediated tumorigenesis.


Asunto(s)
Genes ras , Neoplasias Pancreáticas , Humanos , Sistema de Señalización de MAP Quinasas , Proteómica , Fosfoproteínas/metabolismo , ARN Ribosómico/metabolismo , ARN/metabolismo , Transformación Celular Neoplásica/genética , Ribosomas/genética , Ribosomas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Nucleolina
2.
Nat Methods ; 21(3): 423-434, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38374261

RESUMEN

Different regions of RNA molecules can often engage in specific interactions with distinct RNA-binding proteins (RBPs), giving rise to diverse modalities of RNA regulation and function. However, there are currently no methods for unbiased identification of RBPs that interact with specific RNA regions in living cells and under endogenous settings. Here we introduce TREX (targeted RNase H-mediated extraction of crosslinked RBPs)-a highly sensitive approach for identifying proteins that directly bind to specific RNA regions in living cells. We demonstrate that TREX outperforms existing methods in identifying known interactors of U1 snRNA, and reveals endogenous region-specific interactors of NORAD long noncoding RNA. Using TREX, we generated a comprehensive region-by-region interactome for 45S rRNA, uncovering both established and previously unknown interactions that regulate ribosome biogenesis. With its applicability to different cell types, TREX is an RNA-centric tool for unbiased positional mapping of endogenous RNA-protein interactions in living cells.


Asunto(s)
Proteínas de Unión al ARN , ARN , ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
3.
Development ; 149(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35723257

RESUMEN

Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.


Asunto(s)
Células Endoteliales , Factores de Transcripción , Animales , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Ratones , Neovascularización Fisiológica/genética , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitinas/metabolismo
4.
Development ; 147(4)2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32054660

RESUMEN

La-related protein 6 (Larp6) is a conserved RNA-binding protein found across eukaryotes that has been suggested to regulate collagen biogenesis, muscle development, ciliogenesis, and various aspects of cell proliferation and migration. Zebrafish have two Larp6 family genes: larp6a and larp6b Viable and fertile single and double homozygous larp6a and larp6b zygotic mutants revealed no defects in muscle structure, and were indistinguishable from heterozygous or wild-type siblings. However, larp6a mutant females produced eggs with chorions that failed to elevate fully and were fragile. Eggs from larp6b single mutant females showed minor chorion defects, but chorions from eggs laid by larp6a;larp6b double mutant females were more defective than those from larp6a single mutants. Electron microscopy revealed defective chorionogenesis during oocyte development. Despite this, maternal zygotic single and double mutants were viable and fertile. Mass spectrometry analysis provided a description of chorion protein composition and revealed significant reductions in a subset of zona pellucida and lectin-type proteins between wild-type and mutant chorions that paralleled the severity of the phenotype. We conclude that Larp6 proteins are required for normal oocyte development, chorion formation and egg activation.


Asunto(s)
Autoantígenos/genética , Autoantígenos/fisiología , Corion/fisiología , Oocitos/fisiología , Ribonucleoproteínas/genética , Ribonucleoproteínas/fisiología , Animales , Movimiento Celular , Proliferación Celular , Colágeno/fisiología , Proteínas del Huevo/fisiología , Femenino , Edición Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma , Genotipo , Heterocigoto , Homocigoto , Lectinas/fisiología , Masculino , Mutación , Oocitos/citología , Oogénesis/fisiología , Fenotipo , Pez Cebra , Zona Pelúcida/fisiología , Antígeno SS-B
5.
Br J Cancer ; 125(5): 699-713, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34172930

RESUMEN

BACKGROUND: Metastasis is a hallmark of cancer and responsible for most cancer deaths. Migrastatics were defined as drugs interfering with all modes of cancer cell invasion and thus cancers' ability to metastasise. First anti-metastatic treatments have recently been approved. METHODS: We used bioinformatic analyses of publicly available melanoma databases. Experimentally, we performed in vitro target validation (including 2.5D cell morphology analysis and mass spectrometric analysis of RhoA binding partners), developed a new traceable spontaneously metastasising murine melanoma model for in vivo validation, and employed histology (haematoxylin/eosin and phospho-myosin II staining) to confirm drug action in harvested tumour tissues. RESULTS: Unbiased and targeted bioinformatic analyses identified the Rho kinase (ROCK)-myosin II pathway and its various components as potentially relevant targets in melanoma. In vitro validation demonstrated redundancy of several RhoGEFs upstream of RhoA and confirmed ROCK as a druggable target downstream of RhoA. The anti-metastatic effects of two ROCK inhibitors were demonstrated through in vivo melanoma metastasis tracking and inhibitor effects also confirmed ex vivo by digital pathology. CONCLUSIONS: We proposed a migrastatic drug development pipeline. As part of the pipeline, we provide a new traceable spontaneous melanoma metastasis model for in vivo quantification of metastasis and anti-metastatic effects by non-invasive imaging.


Asunto(s)
Biología Computacional/métodos , Melanoma/tratamiento farmacológico , Miosina Tipo II/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Masculino , Espectrometría de Masas , Melanoma/metabolismo , Ratones , Metástasis de la Neoplasia , Mapas de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Cell Sci ; 129(24): 4466-4479, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807006

RESUMEN

Directional cell migration involves reorientation of the secretory machinery. However, the molecular mechanisms that control this reorientation are not well characterised. Here, we identify a new Rho effector protein, named FAM65A, which binds to active RHOA, RHOB and RHOC. FAM65A links RHO proteins to Golgi-localising cerebral cavernous malformation-3 protein (CCM3; also known as PDCD10) and its interacting proteins mammalian STE20-like protein kinases 3 and 4 (MST3 and MST4; also known as STK24 and STK26, respectively). Binding of active RHO proteins to FAM65A does not affect the kinase activity of MSTs but results in their relocation from the Golgi in a CCM3-dependent manner. This relocation is crucial for reorientation of the Golgi towards the leading edge and subsequent directional cell migration. Our results reveal a previously unidentified pathway downstream of RHO that regulates the polarity of migrating cells through Golgi reorientation in a FAM65A-, CCM3- and MST3- and MST4-dependent manner.


Asunto(s)
Movimiento Celular , Aparato de Golgi/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Activación Enzimática , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas de la Membrana/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo
7.
PLoS Med ; 13(2): e1001961, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26881778

RESUMEN

BACKGROUND: The intra-tumor diversity of cancer cells is under intense investigation; however, little is known about the heterogeneity of the tumor microenvironment that is key to cancer progression and evolution. We aimed to assess the degree of microenvironmental heterogeneity in breast cancer and correlate this with genomic and clinical parameters. METHODS AND FINDINGS: We developed a quantitative measure of microenvironmental heterogeneity along three spatial dimensions (3-D) in solid tumors, termed the tumor ecosystem diversity index (EDI), using fully automated histology image analysis coupled with statistical measures commonly used in ecology. This measure was compared with disease-specific survival, key mutations, genome-wide copy number, and expression profiling data in a retrospective study of 510 breast cancer patients as a test set and 516 breast cancer patients as an independent validation set. In high-grade (grade 3) breast cancers, we uncovered a striking link between high microenvironmental heterogeneity measured by EDI and a poor prognosis that cannot be explained by tumor size, genomics, or any other data types. However, this association was not observed in low-grade (grade 1 and 2) breast cancers. The prognostic value of EDI was superior to known prognostic factors and was enhanced with the addition of TP53 mutation status (multivariate analysis test set, p = 9 × 10-4, hazard ratio = 1.47, 95% CI 1.17-1.84; validation set, p = 0.0011, hazard ratio = 1.78, 95% CI 1.26-2.52). Integration with genome-wide profiling data identified losses of specific genes on 4p14 and 5q13 that were enriched in grade 3 tumors with high microenvironmental diversity that also substratified patients into poor prognostic groups. Limitations of this study include the number of cell types included in the model, that EDI has prognostic value only in grade 3 tumors, and that our spatial heterogeneity measure was dependent on spatial scale and tumor size. CONCLUSIONS: To our knowledge, this is the first study to couple unbiased measures of microenvironmental heterogeneity with genomic alterations to predict breast cancer clinical outcome. We propose a clinically relevant role of microenvironmental heterogeneity for advanced breast tumors, and highlight that ecological statistics can be translated into medical advances for identifying a new type of biomarker and, furthermore, for understanding the synergistic interplay of microenvironmental heterogeneity with genomic alterations in cancer cells.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , ADN de Neoplasias/genética , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Estadificación de Neoplasias , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Adulto Joven
8.
Elife ; 132024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38597390

RESUMEN

Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.


Asunto(s)
Empalme Alternativo , Exones , Neuronas , Proteína de Unión al Tracto de Polipirimidina , Proteínas Serina-Treonina Quinasas , Animales , Humanos , Ratones , Exones/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Fosforilación , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo
9.
Oncogene ; 43(21): 1608-1619, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565943

RESUMEN

Cancer cells employ adaptive mechanisms to survive various stressors, including genotoxic drugs. Understanding the factors promoting survival is crucial for developing effective treatments. In this study, we unveil a previously unexplored long non-coding RNA, JUNI (JUN-DT, LINC01135), which is upregulated by genotoxic drugs through the activation of stress-activated MAPKs, JNK, and p38 and consequently exerts positive control over the expression of its adjacent gene product c-Jun, a well-known oncoprotein, which transduces signals to multiple transcriptional outputs. JUNI regulates cellular migration and has a crucial role in conferring cellular resistance to chemotherapeutic drugs or UV radiation. Depletion of JUNI markedly increases the sensitivity of cultured cells and spheroids to chemotherapeutic agents. We identified 57 proteins interacting with JUNI. The activity of one of them the MAPK phosphatase and inhibitor, DUSP14, is counteracted by JUNI, thereby, facilitating efficient JNK phosphorylation and c-Jun induction when cells are exposed to UV radiation. The antagonistic interplay with DUSP14 contributes not only to c-Jun induction but also augments the survival of UV-exposed cells. In summary, we introduce JUNI as a novel stress-inducible regulator of c-Jun, positioning it as a potential target for enhancing the sensitivity of cancer cells to chemotherapy.


Asunto(s)
Movimiento Celular , Supervivencia Celular , Fosfatasas de Especificidad Dual , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/genética , Movimiento Celular/genética , Supervivencia Celular/efectos de la radiación , Supervivencia Celular/genética , Supervivencia Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Línea Celular Tumoral , Rayos Ultravioleta/efectos adversos , Sistema de Señalización de MAP Quinasas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo
10.
Nat Cell Biol ; 25(6): 892-903, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37156909

RESUMEN

Cancer cells often co-opt post-transcriptional regulatory mechanisms to achieve pathologic expression of gene networks that drive metastasis. Translational control is a major regulatory hub in oncogenesis; however, its effects on cancer progression remain poorly understood. Here, to address this, we used ribosome profiling to compare genome-wide translation efficiencies of poorly and highly metastatic breast cancer cells and patient-derived xenografts. We developed dedicated regression-based methods to analyse ribosome profiling and alternative polyadenylation data, and identified heterogeneous nuclear ribonucleoprotein C (HNRNPC) as a translational controller of a specific mRNA regulon. We found that HNRNPC is downregulated in highly metastatic cells, which causes HNRNPC-bound mRNAs to undergo 3' untranslated region lengthening and, subsequently, translational repression. We showed that modulating HNRNPC expression impacts the metastatic capacity of breast cancer cells in xenograft mouse models. In addition, the reduced expression of HNRNPC and its regulon is associated with the worse prognosis in breast cancer patient cohorts.


Asunto(s)
Neoplasias de la Mama , Procesamiento Postranscripcional del ARN , Humanos , Animales , Ratones , Femenino , Neoplasias de la Mama/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
STAR Protoc ; 2(2): 100462, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33912849

RESUMEN

Actin-rich protrusions are membrane extensions generated by actin polymerization that drive mesenchymal-like cell migration. Characterization of protrusions proteome is crucial for understanding their function. We present a complete step-by-step protocol based on microporous filter-based fractionation of protrusive cellular domains coupled with sample preparation for quantitative proteomics, mass spectrometric data acquisition, and data analysis. This protocol enables purification, quantification, and analysis of the distribution of proteins present in protrusions and cell bodies. For complete details on the use and execution of this protocol, please refer to Dermit et al. (2020).


Asunto(s)
Cuerpo Celular , Extensiones de la Superficie Celular , Proteómica , Cuerpo Celular/química , Cuerpo Celular/metabolismo , Línea Celular Tumoral , Extensiones de la Superficie Celular/química , Extensiones de la Superficie Celular/metabolismo , Humanos
12.
Cell Death Dis ; 12(11): 1075, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764236

RESUMEN

An early event in lung oncogenesis is loss of the tumour suppressor gene LIMD1 (LIM domains containing 1); this encodes a scaffold protein, which suppresses tumorigenesis via a number of different mechanisms. Approximately 45% of non-small cell lung cancers (NSCLC) are deficient in LIMD1, yet this subtype of NSCLC has been overlooked in preclinical and clinical investigations. Defining therapeutic targets in these LIMD1 loss-of-function patients is difficult due to a lack of 'druggable' targets, thus alternative approaches are required. To this end, we performed the first drug repurposing screen to identify compounds that confer synthetic lethality with LIMD1 loss in NSCLC cells. PF-477736 was shown to selectively target LIMD1-deficient cells in vitro through inhibition of multiple kinases, inducing cell death via apoptosis. Furthermore, PF-477736 was effective in treating LIMD1-/- tumours in subcutaneous xenograft models, with no significant effect in LIMD1+/+ cells. We have identified a novel drug tool with significant preclinical characterisation that serves as an excellent candidate to explore and define LIMD1-deficient cancers as a new therapeutic subgroup of critical unmet need.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas con Dominio LIM/deficiencia , Neoplasias Pulmonares/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Prueba de Estudio Conceptual , Transfección
13.
Curr Biol ; 31(7): 1403-1416.e7, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33592190

RESUMEN

Bidirectional communication between cells and their surrounding environment is critical in both normal and pathological settings. Extracellular vesicles (EVs), which facilitate the horizontal transfer of molecules between cells, are recognized as an important constituent of cell-cell communication. In cancer, alterations in EV secretion contribute to the growth and metastasis of tumor cells. However, the mechanisms underlying these changes remain largely unknown. Here, we show that centrosome amplification is associated with and sufficient to promote small extracellular vesicle (SEV) secretion in pancreatic cancer cells. This is a direct result of lysosomal dysfunction, caused by increased reactive oxygen species (ROS) downstream of extra centrosomes. We propose that defects in lysosome function could promote multivesicular body fusion with the plasma membrane, thereby enhancing SEV secretion. Furthermore, we find that SEVs secreted in response to amplified centrosomes are functionally distinct and activate pancreatic stellate cells (PSCs). These activated PSCs promote the invasion of pancreatic cancer cells in heterotypic 3D cultures. We propose that SEVs secreted by cancer cells with amplified centrosomes influence the bidirectional communication between the tumor cells and the surrounding stroma to promote malignancy.


Asunto(s)
Centrosoma , Vesículas Extracelulares , Lisosomas , Animales , Vesículas Extracelulares/metabolismo , Humanos , Ratones , Cuerpos Multivesiculares , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
14.
Sci Signal ; 14(691)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34257105

RESUMEN

Synaptic plasticity involves structural modifications in dendritic spines that are modulated by local protein synthesis and actin remodeling. Here, we investigated the molecular mechanisms that connect synaptic stimulation to these processes. We found that the phosphorylation of isoform-specific sites in eEF1A2-an essential translation elongation factor in neurons-is a key modulator of structural plasticity in dendritic spines. Expression of a nonphosphorylatable eEF1A2 mutant stimulated mRNA translation but reduced actin dynamics and spine density. By contrast, a phosphomimetic eEF1A2 mutant exhibited decreased association with F-actin and was inactive as a translation elongation factor. Activation of metabotropic glutamate receptor signaling triggered transient dissociation of eEF1A2 from its regulatory guanine exchange factor (GEF) protein in dendritic spines in a phosphorylation-dependent manner. We propose that eEF1A2 establishes a cross-talk mechanism that coordinates translation and actin dynamics during spine remodeling.


Asunto(s)
Actinas , Espinas Dendríticas , Factor 1 de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Citoesqueleto de Actina , Actinas/genética , Plasticidad Neuronal , Neuronas
15.
Science ; 372(6543)2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33986153

RESUMEN

Aberrant alternative splicing is a hallmark of cancer, yet the underlying regulatory programs that control this process remain largely unknown. Here, we report a systematic effort to decipher the RNA structural code that shapes pathological splicing during breast cancer metastasis. We discovered a previously unknown structural splicing enhancer that is enriched near cassette exons with increased inclusion in highly metastatic cells. We show that the spliceosomal protein small nuclear ribonucleoprotein polypeptide A' (SNRPA1) interacts with these enhancers to promote cassette exon inclusion. This interaction enhances metastatic lung colonization and cancer cell invasion, in part through SNRPA1-mediated regulation of PLEC alternative splicing, which can be counteracted by splicing modulating morpholinos. Our findings establish a noncanonical regulatory role for SNRPA1 as a prometastatic splicing enhancer in breast cancer.


Asunto(s)
Empalme Alternativo , Neoplasias de la Mama/patología , Metástasis de la Neoplasia/genética , ARN/genética , ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Algoritmos , Animales , Sitios de Unión , Neoplasias de la Mama/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Exones , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Conformación de Ácido Nucleico , Plectina/genética , Unión Proteica , Interferencia de ARN , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/metabolismo , RNA-Seq , Ribonucleoproteína Nuclear Pequeña U2/genética , Programas Informáticos , Empalmosomas/metabolismo , Proteínas Supresoras de Tumor/genética
16.
Dev Cell ; 55(3): 298-313.e10, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33171110

RESUMEN

Translation of ribosomal protein-coding mRNAs (RP-mRNAs) constitutes a key step in ribosome biogenesis, but the mechanisms that modulate RP-mRNA translation in coordination with other cellular processes are poorly defined. Here, we show that subcellular localization of RP-mRNAs acts as a key regulator of their translation during cell migration. As cells migrate into their surroundings, RP-mRNAs localize to the actin-rich cell protrusions. This localization is mediated by La-related protein 6 (LARP6), an RNA-binding protein that is enriched in protrusions. Protrusions act as hotspots of translation for RP-mRNAs, enhancing RP synthesis, ribosome biogenesis, and the overall protein synthesis in migratory cells. In human breast carcinomas, epithelial-to-mesenchymal transition (EMT) upregulates LARP6 expression to enhance protein synthesis and support invasive growth. Our findings reveal LARP6-mediated mRNA localization as a key regulator of ribosome biogenesis during cell migration and demonstrate a role for this process in cancer progression downstream of EMT.


Asunto(s)
Movimiento Celular , Biogénesis de Organelos , Transporte de ARN , Ribosomas/metabolismo , Autoantígenos/metabolismo , Proliferación Celular , Extensiones de la Superficie Celular/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Fracciones Subcelulares/metabolismo , Transcriptoma/genética , Antígeno SS-B
17.
Cancer Discov ; 10(9): 1410-1423, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32513775

RESUMEN

Identifying master regulators that drive pathologic gene expression is a key challenge in precision oncology. Here, we have developed an analytic framework, named PRADA, that identifies oncogenic RNA-binding proteins through the systematic detection of coordinated changes in their target regulons. Application of this approach to data collected from clinical samples, patient-derived xenografts, and cell line models of colon cancer metastasis revealed the RNA-binding protein RBMS1 as a suppressor of colon cancer progression. We observed that silencing RBMS1 results in increased metastatic capacity in xenograft mouse models, and that restoring its expression blunts metastatic liver colonization. We have found that RBMS1 functions as a posttranscriptional regulator of RNA stability by directly binding its target mRNAs. Together, our findings establish a role for RBMS1 as a previously unknown regulator of RNA stability and as a suppressor of colon cancer metastasis with clinical utility for risk stratification of patients. SIGNIFICANCE: By applying a new analytic approach to transcriptomic data from clinical samples and models of colon cancer progression, we have identified RBMS1 as a suppressor of metastasis and as a post-transcriptional regulator of RNA stability. Notably, RBMS1 silencing and downregulation of its targets are negatively associated with patient survival.See related commentary by Carter, p. 1261.This article is highlighted in the In This Issue feature, p. 1241.


Asunto(s)
Neoplasias del Colon/patología , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Colon/patología , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/genética , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Humanos , Mucosa Intestinal/patología , Neoplasias Hepáticas/secundario , Masculino , Ratones , Estadificación de Neoplasias , Estabilidad del ARN/genética , Proteínas de Unión al ARN/genética , RNA-Seq , Regulón , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Anal Chem ; 80(17): 6650-7, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18683950

RESUMEN

We demonstrate a strategy employing collision-induced dissociation for phosphopeptide discovery, followed by targeted electron capture dissociation (ECD) for site localization. The high mass accuracy and low background noise of the ECD mass spectra allow facile sequencing of coeluting isobaric phosphopeptides, with up to two isobaric phosphopeptides sequenced from a single mass spectrum. In contrast to the previously described neutral loss dependent ECD method, targeted ECD allows analysis of both phosphotyrosine peptides and lower abundance phosphopeptides. The approach was applied to phosphorylation analysis of human Sprouty2, a regulator of receptor tyrosine kinase signaling. Fifteen sites of phosphorylation were identified, 11 of which are novel.


Asunto(s)
Electrones , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Línea Celular , Cromatografía Liquida , Humanos , Espectrometría de Masas , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación , Sensibilidad y Especificidad
19.
J Clin Invest ; 128(4): 1671-1687, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29447131

RESUMEN

Synthetic lethality is an efficient mechanism-based approach to selectively target DNA repair defects. Excision repair cross-complementation group 1 (ERCC1) deficiency is frequently found in non-small-cell lung cancer (NSCLC), making this DNA repair protein an attractive target for exploiting synthetic lethal approaches in the disease. Using unbiased proteomic and metabolic high-throughput profiling on a unique in-house-generated isogenic model of ERCC1 deficiency, we found marked metabolic rewiring of ERCC1-deficient populations, including decreased levels of the metabolite NAD+ and reduced expression of the rate-limiting NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT). We also found reduced NAMPT expression in NSCLC samples with low levels of ERCC1. These metabolic alterations were a primary effect of ERCC1 deficiency, and caused selective exquisite sensitivity to small-molecule NAMPT inhibitors, both in vitro - ERCC1-deficient cells being approximately 1,000 times more sensitive than ERCC1-WT cells - and in vivo. Using transmission electronic microscopy and functional metabolic studies, we found that ERCC1-deficient cells harbor mitochondrial defects. We propose a model where NAD+ acts as a regulator of ERCC1-deficient NSCLC cell fitness. These findings open therapeutic opportunities that exploit a yet-undescribed nuclear-mitochondrial synthetic lethal relationship in NSCLC models, and highlight the potential for targeting DNA repair/metabolic crosstalks for cancer therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Reparación del ADN , Neoplasias Pulmonares/metabolismo , NAD/biosíntesis , Neoplasias Experimentales/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Citocinas/genética , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , NAD/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Nicotinamida Fosforribosiltransferasa/genética , Nicotinamida Fosforribosiltransferasa/metabolismo
20.
Methods Mol Biol ; 1636: 337-352, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28730490

RESUMEN

A major challenge in systems biology is comprehensive mapping of protein interaction networks. Crucially, such interactions are often dynamic in nature, necessitating methods that can rapidly mine the interactome across varied conditions and treatments to reveal change in the interaction networks. Recently, we described a fast mass spectrometry-based method to reveal functional interactions in mammalian cells on a global scale, by revealing spatial colocalizations between proteins (COLA) (Mardakheh et al., Mol Biosyst 13:92-105, 2017). As protein localization and function are inherently linked, significant colocalization between two proteins is a strong indication for their functional interaction. COLA uses rapid complete subcellular fractionation, coupled with quantitative proteomics to generate a subcellular localization profile for each protein quantified by the mass spectrometer. Robust clustering is then applied to reveal significant similarities in protein localization profiles, indicative of colocalization.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , Proteoma , Proteómica/métodos , Fraccionamiento Celular/métodos , Cromatografía Liquida , Análisis por Conglomerados , Biología Computacional/métodos , Espectrometría de Masas , Unión Proteica , Transporte de Proteínas , Coloración y Etiquetado , Estadística como Asunto , Flujo de Trabajo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA