Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunity ; 57(1): 153-170.e6, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38159571

RESUMEN

The dominant risk factors for late-onset Alzheimer's disease (AD) are advanced age and the APOE4 genetic variant. To examine how these factors alter neuroimmune function, we generated an integrative, longitudinal single-cell atlas of brain immune cells in AD model mice bearing the three common human APOE alleles. Transcriptomic and chromatin accessibility analyses identified a reactive microglial population defined by the concomitant expression of inflammatory signals and cell-intrinsic stress markers whose frequency increased with age and APOE4 burden. An analogous population was detectable in the brains of human AD patients, including in the cortical tissue, using multiplexed spatial transcriptomics. This population, which we designate as terminally inflammatory microglia (TIM), exhibited defects in amyloid-ß clearance and altered cell-cell communication during aducanumab treatment. TIM may represent an exhausted-like state for inflammatory microglia in the AD milieu that contributes to AD risk and pathology in APOE4 carriers and the elderly, thus presenting a potential therapeutic target for AD.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Anciano , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/genética , Encéfalo/metabolismo , Genotipo , Microglía
2.
Cell ; 172(4): 825-840.e18, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29336888

RESUMEN

Therapeutic harnessing of adaptive immunity via checkpoint inhibition has transformed the treatment of many cancers. Despite unprecedented long-term responses, most patients do not respond to these therapies. Immunotherapy non-responders often harbor high levels of circulating myeloid-derived suppressor cells (MDSCs)-an immunosuppressive innate cell population. Through genetic and pharmacological approaches, we uncovered a pathway governing MDSC abundance in multiple cancer types. Therapeutic liver-X nuclear receptor (LXR) agonism reduced MDSC abundance in murine models and in patients treated in a first-in-human dose escalation phase 1 trial. MDSC depletion was associated with activation of cytotoxic T lymphocyte (CTL) responses in mice and patients. The LXR transcriptional target ApoE mediated these effects in mice, where LXR/ApoE activation therapy elicited robust anti-tumor responses and also enhanced T cell activation during various immune-based therapies. We implicate the LXR/ApoE axis in the regulation of innate immune suppression and as a target for enhancing the efficacy of cancer immunotherapy in patients.


Asunto(s)
Apolipoproteínas E/inmunología , Inmunidad Innata , Receptores X del Hígado/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias Experimentales/inmunología , Animales , Apolipoproteínas E/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Femenino , Receptores X del Hígado/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células Supresoras de Origen Mieloide/patología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Cell ; 84(10): 1819-1821, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38759621

RESUMEN

In this issue of Molecular Cell, Yang et al.1 find that arginine-to-cysteine substitutants are enriched in a subset of lung cancer proteomes, potentiated by arginine deprivation, and promote resistance to chemotherapy.


Asunto(s)
Arginina , Cisteína , Neoplasias Pulmonares , Proteoma , Humanos , Cisteína/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Arginina/metabolismo , Proteoma/metabolismo , Resistencia a Antineoplásicos/genética
4.
Immunity ; 55(4): 580-582, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417670

RESUMEN

Interferon signaling mediates resistance to immune checkpoint blockade therapy, but the underlying mechanisms are poorly understood. In this issue of Immunity, Cucolo et al. identify RIPK1 as an interferon-stimulated gene with potent effects on cell extrinsic and intrinsic immunotherapy resistance.


Asunto(s)
Resistencia a Antineoplásicos , Inmunoterapia , Neoplasias , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Humanos , Factores Inmunológicos , Interferones
5.
Cell ; 165(6): 1416-1427, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259150

RESUMEN

Transfer RNAs (tRNAs) are primarily viewed as static contributors to gene expression. By developing a high-throughput tRNA profiling method, we find that specific tRNAs are upregulated in human breast cancer cells as they gain metastatic activity. Through loss-of-function, gain-of-function, and clinical-association studies, we implicate tRNAGluUUC and tRNAArgCCG as promoters of breast cancer metastasis. Upregulation of these tRNAs enhances stability and ribosome occupancy of transcripts enriched for their cognate codons. Specifically, tRNAGluUUC promotes metastatic progression by directly enhancing EXOSC2 expression and enhancing GRIPAP1-constituting an "inducible" pathway driven by a tRNA. The cellular proteomic shift toward a pro-metastatic state mirrors global tRNA shifts, allowing for cell-state and cell-type transgene expression optimization through codon content quantification. TRNA modulation represents a mechanism by which cells achieve altered expression of specific transcripts and proteins. TRNAs are thus dynamic regulators of gene expression and the tRNA codon landscape can causally and specifically impact disease progression.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , ARN de Transferencia/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Codón , Progresión de la Enfermedad , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Pulmón/patología , Ratones , Invasividad Neoplásica/genética , Micrometástasis de Neoplasia/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Unión al ARN/genética , Secuencias Reguladoras de Ácido Ribonucleico , Ribosomas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell ; 161(4): 790-802, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25957686

RESUMEN

Upon exposure to stress, tRNAs are enzymatically cleaved, yielding distinct classes of tRNA-derived fragments (tRFs), yielding distinct classes of tRFs. We identify a novel class of tRFs derived from tRNA(Glu), tRNA(Asp), tRNA(Gly), and tRNA(Tyr) that, upon induction, suppress the stability of multiple oncogenic transcripts in breast cancer cells by displacing their 3' untranslated regions (UTRs) from the RNA-binding protein YBX1. This mode of post-transcriptional silencing is sequence specific, as these fragments all share a common motif that matches the YBX1 recognition sequence. Loss-of-function and gain-of-function studies, using anti-sense locked-nucleic acids (LNAs) and synthetic RNA mimetics, respectively, revealed that these fragments suppress growth under serum-starvation, cancer cell invasion, and metastasis by breast cancer cells. Highly metastatic cells evade this tumor-suppressive pathway by attenuating the induction of these tRFs. Our findings reveal a tumor-suppressive role for specific tRNA-derived fragments and describe a molecular mechanism for their action. This transcript displacement-based mechanism may generalize to other tRNA, ribosomal-RNA, and sno-RNA fragments.


Asunto(s)
Neoplasias de la Mama/patología , ARN Pequeño no Traducido/metabolismo , Proteína 1 de Unión a la Caja Y/antagonistas & inhibidores , Proteína 1 de Unión a la Caja Y/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Células HEK293 , Humanos , Metástasis de la Neoplasia , Oligonucleótidos/farmacología , ARN Pequeño no Traducido/análisis , ARN Pequeño no Traducido/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Análisis de Secuencia de ARN
7.
Cell ; 162(6): 1299-308, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26321680

RESUMEN

N(6)-methyladenosine (m(6)A) is the most abundant internal modification of messenger RNA. While the presence of m(6)A on transcripts can impact nuclear RNA fates, a reader of this mark that mediates processing of nuclear transcripts has not been identified. We find that the RNA-binding protein HNRNPA2B1 binds m(6)A-bearing RNAs in vivo and in vitro and its biochemical footprint matches the m(6)A consensus motif. HNRNPA2B1 directly binds a set of nuclear transcripts and elicits similar alternative splicing effects as the m(6)A writer METTL3. Moreover, HNRNPA2B1 binds to m(6)A marks in a subset of primary miRNA transcripts, interacts with the microRNA Microprocessor complex protein DGCR8, and promotes primary miRNA processing. Also, HNRNPA2B1 loss and METTL3 depletion cause similar processing defects for these pri-miRNA precursors. We propose HNRNPA2B1 to be a nuclear reader of the m(6)A mark and to mediate, in part, this mark's effects on primary microRNA processing and alternative splicing. PAPERCLIP.


Asunto(s)
Adenosina/análogos & derivados , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Procesamiento Postranscripcional del ARN , Adenosina/metabolismo , Empalme Alternativo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Metilación , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcriptoma
8.
Cell ; 160(3): 393-406, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25601461

RESUMEN

Colorectal cancer primarily metastasizes to the liver and globally kills over 600,000 people annually. By functionally screening 661 microRNAs (miRNAs) in parallel during liver colonization, we have identified miR-551a and miR-483 as robust endogenous suppressors of liver colonization and metastasis. These miRNAs convergently target creatine kinase, brain-type (CKB), which phosphorylates the metabolite creatine, to generate phosphocreatine. CKB is released into the extracellular space by metastatic cells encountering hepatic hypoxia and catalyzes production of phosphocreatine, which is imported through the SLC6A8 transporter and used to generate ATP­fueling metastatic survival. Combinatorial therapeutic viral delivery of miR-551a and miR-483-5p through single-dose adeno-associated viral (AAV) delivery significantly suppressed colon cancer metastasis, as did CKB inhibition with a small-molecule inhibitor. Importantly, human liver metastases express higher CKB and SLC6A8 levels and reduced miR-551a/miR-483 levels relative to primary tumors. We identify the extracellular space as an important compartment for malignant energetic catalysis and therapeutic targeting.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Hepáticas/secundario , MicroARNs/metabolismo , Metástasis de la Neoplasia/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Forma BB de la Creatina-Quinasa/metabolismo , Metabolismo Energético , Matriz Extracelular , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo
9.
Mol Cell ; 82(14): 2604-2617.e8, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35654044

RESUMEN

Stress-induced cleavage of transfer RNAs (tRNAs) into tRNA-derived fragments (tRFs) occurs across organisms from yeast to humans; yet, its mechanistic underpinnings and pathological consequences remain poorly defined. Small RNA profiling revealed increased abundance of a cysteine tRNA fragment (5'-tRFCys) during breast cancer metastatic progression. 5'-tRFCys was required for efficient breast cancer metastatic lung colonization and cancer cell survival. We identified Nucleolin as the direct binding partner of 5'-tRFCys. 5'-tRFCys promoted the oligomerization of Nucleolin and its bound metabolic transcripts Mthfd1l and Pafah1b1 into a higher-order transcript stabilizing ribonucleoprotein complex, which protected these transcripts from exonucleolytic degradation. Consistent with this, Mthfd1l and Pafah1b1 mediated pro-metastatic and metabolic effects downstream of 5'-tRFCys-impacting folate, one-carbon, and phosphatidylcholine metabolism. Our findings reveal that a tRF can promote oligomerization of an RNA-binding protein into a transcript stabilizing ribonucleoprotein complex, thereby driving specific metabolic pathways underlying cancer progression.


Asunto(s)
Neoplasias de la Mama , ARN de Transferencia , Neoplasias de la Mama/genética , Femenino , Humanos , Fosfoproteínas , ARN Mensajero/genética , ARN de Transferencia/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/genética , Nucleolina
10.
Cell ; 156(5): 986-1001, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581497

RESUMEN

Melanoma metastasis is a devastating outcome lacking an effective preventative therapeutic. We provide pharmacologic, molecular, and genetic evidence establishing the liver-X nuclear hormone receptor (LXR) as a therapeutic target in melanoma. Oral administration of multiple LXR agonists suppressed melanoma invasion, angiogenesis, tumor progression, and metastasis. Molecular and genetic experiments revealed these effects to be mediated by LXRß, which elicits these outcomes through transcriptional induction of tumoral and stromal apolipoprotein-E (ApoE). LXRß agonism robustly suppressed tumor growth and metastasis across a diverse mutational spectrum of melanoma lines. LXRß targeting significantly prolonged animal survival, suppressed the progression of established metastases, and inhibited brain metastatic colonization. Importantly, LXRß activation displayed melanoma-suppressive cooperativity with the frontline regimens dacarbazine, B-Raf inhibition, and the anti-CTLA-4 antibody and robustly inhibited melanomas that had acquired resistance to B-Raf inhibition or dacarbazine. We present a promising therapeutic approach that uniquely acts by transcriptionally activating a metastasis suppressor gene.


Asunto(s)
Melanoma/tratamiento farmacológico , Melanoma/secundario , Metástasis de la Neoplasia/tratamiento farmacológico , Receptores Nucleares Huérfanos/agonistas , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Benzoatos/administración & dosificación , Bencilaminas/administración & dosificación , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Hidrocarburos Fluorados/administración & dosificación , Receptores X del Hígado , Melanoma/patología , Ratones , Metástasis de la Neoplasia/patología , Transducción de Señal , Neoplasias Cutáneas/patología , Sulfonamidas/administración & dosificación , Transcripción Genética
11.
Nature ; 611(7935): 346-351, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36130725

RESUMEN

Clinical outcomes of severe acute respiratory syndrome 2 (SARS-CoV-2) infection are highly heterogeneous, ranging from asymptomatic infection to lethal coronavirus disease 2019 (COVID-19). The factors underlying this heterogeneity remain insufficiently understood. Genetic association studies have suggested that genetic variants contribute to the heterogeneity of COVID-19 outcomes, but the underlying potential causal mechanisms are insufficiently understood. Here we show that common variants of the apolipoprotein E (APOE) gene, homozygous in approximately 3% of the world's population1 and associated with Alzheimer's disease, atherosclerosis and anti-tumour immunity2-5, affect COVID-19 outcome in a mouse model that recapitulates increased susceptibility conferred by male sex and advanced age. Mice bearing the APOE2 or APOE4 variant exhibited rapid disease progression and poor survival outcomes relative to mice bearing the most prevalent APOE3 allele. APOE2 and APOE4 mice exhibited increased viral loads as well as suppressed adaptive immune responses early after infection. In vitro assays demonstrated increased infection in the presence of APOE2 and APOE4 relative to APOE3, indicating that differential outcomes are mediated by differential effects of APOE variants on both viral infection and antiviral immunity. Consistent with these in vivo findings in mice, our results also show that APOE genotype is associated with survival in patients infected with SARS-CoV-2 in the UK Biobank (candidate variant analysis, P = 2.6 × 10-7). Our findings suggest APOE genotype to partially explain the heterogeneity of COVID-19 outcomes and warrant prospective studies to assess APOE genotyping as a means of identifying patients at high risk for adverse outcomes.


Asunto(s)
Apolipoproteínas E , COVID-19 , Genética Humana , Ratones Transgénicos , SARS-CoV-2 , Animales , Humanos , Masculino , Ratones , Apolipoproteína E2/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , COVID-19/genética , COVID-19/mortalidad , COVID-19/virología , Ratones Transgénicos/genética , Ratones Transgénicos/virología , Estudios Prospectivos , SARS-CoV-2/patogenicidad , Modelos Animales de Enfermedad
12.
Cell ; 151(5): 1068-82, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23142051

RESUMEN

Through in vivo selection of human cancer cell populations, we uncover a convergent and cooperative miRNA network that drives melanoma metastasis. We identify miR-1908, miR-199a-5p, and miR-199a-3p as endogenous promoters of metastatic invasion, angiogenesis, and colonization in melanoma. These miRNAs convergently target apolipoprotein E (ApoE) and the heat shock factor DNAJA4. Cancer-secreted ApoE suppresses invasion and metastatic endothelial recruitment (MER) by engaging melanoma cell LRP1 and endothelial cell LRP8 receptors, respectively, while DNAJA4 promotes ApoE expression. Expression levels of these miRNAs and ApoE correlate with human metastatic progression outcomes. Treatment of cells with locked nucleic acids (LNAs) targeting these miRNAs inhibits metastasis to multiple organs, and therapeutic delivery of these LNAs strongly suppresses melanoma metastasis. We thus identify miRNAs with dual cell-intrinsic/cell-extrinsic roles in cancer, reveal convergent cooperativity in a metastatic miRNA network, identify ApoE as an anti-angiogenic and metastasis-suppressive factor, and uncover multiple prognostic miRNAs with synergistic combinatorial therapeutic potential in melanoma.


Asunto(s)
Apolipoproteínas E/metabolismo , Melanoma/genética , MicroARNs/metabolismo , Metástasis de la Neoplasia/genética , Neovascularización Patológica/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Oligonucleótidos/farmacología
13.
Nature ; 586(7828): 299-304, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32999457

RESUMEN

Blood vessels support tumours by providing nutrients and oxygen, while also acting as conduits for the dissemination of cancer1. Here we use mouse models of breast and lung cancer to investigate whether endothelial cells also have active 'instructive' roles in the dissemination of cancer. We purified genetically tagged endothelial ribosomes and their associated transcripts from highly and poorly metastatic tumours. Deep sequencing revealed that metastatic tumours induced expression of the axon-guidance gene Slit2 in endothelium, establishing differential expression between the endothelial (high Slit2 expression) and tumoural (low Slit2 expression) compartments. Endothelial-derived SLIT2 protein and its receptor ROBO1 promoted the migration of cancer cells towards endothelial cells and intravasation. Deleting endothelial Slit2 suppressed metastatic dissemination in mouse models of breast and lung cancer. Conversely, deletion of tumoural Slit2 enhanced metastatic progression. We identified double-stranded RNA derived from tumour cells as an upstream signal that induces expression of endothelial SLIT2 by acting on the RNA-sensing receptor TLR3. Accordingly, a set of endogenous retroviral element RNAs were upregulated in metastatic cells and detected extracellularly. Thus, cancer cells co-opt innate RNA sensing to induce a chemotactic signalling pathway in endothelium that drives intravasation and metastasis. These findings reveal that endothelial cells have a direct instructive role in driving metastatic dissemination, and demonstrate that a single gene (Slit2) can promote or suppress cancer progression depending on its cellular source.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Endotelio/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Proteínas del Tejido Nervioso/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Quimiotaxis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Metástasis de la Neoplasia/genética , Proteínas del Tejido Nervioso/genética , ARN Bicatenario , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Células Tumorales Cultivadas , Proteínas Roundabout
14.
EMBO J ; 40(2): e106696, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33346941

RESUMEN

Eukaryotic transfer RNAs can become selectively fragmented upon various stresses, generating tRNA-derived small RNA fragments. Such fragmentation has been reported to impact a small fraction of the tRNA pool and thus presumed to not directly impact translation. We report that oxidative stress can rapidly generate tyrosine-tRNAGUA fragments in human cells-causing significant depletion of the precursor tRNA. Tyrosine-tRNAGUA depletion impaired translation of growth and metabolic genes enriched in cognate tyrosine codons. Depletion of tyrosine tRNAGUA or its translationally regulated targets USP3 and SCD repressed proliferation-revealing a dedicated tRNA-regulated growth-suppressive pathway for oxidative stress response. Tyrosine fragments are generated in a DIS3L2 exoribonuclease-dependent manner and inhibit hnRNPA1-mediated transcript destabilization. Moreover, tyrosine fragmentation is conserved in C. elegans. Thus, tRNA fragmentation can coordinately generate trans-acting small RNAs and functionally deplete a tRNA. Our findings reveal the existence of an underlying adaptive codon-based regulatory response inherent to the genetic code.


Asunto(s)
Codón/genética , Biosíntesis de Proteínas/genética , ARN de Transferencia/genética , Tirosina/genética , Animales , Caenorhabditis elegans/genética , Línea Celular , Proliferación Celular/genética , Células HEK293 , Humanos , Estrés Oxidativo/genética , Proteasas Ubiquitina-Específicas/genética
15.
Genes Dev ; 30(4): 386-98, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26883358

RESUMEN

Post-transcriptional deregulation is a defining feature of metastatic cancer. While many microRNAs have been implicated as regulators of metastatic progression, less is known about the roles and mechanisms of RNA-binding proteins in this process. We identified muscleblind-like 1 (MBNL1), a gene implicated in myotonic dystrophy, as a robust suppressor of multiorgan breast cancer metastasis. MBNL1 binds the 3' untranslated regions (UTRs) of DBNL (drebrin-like protein) and TACC1 (transforming acidic coiled-coil containing protein 1)-two genes that we implicate as metastasis suppressors. By enhancing the stability of these genes' transcripts, MBNL1 suppresses cell invasiveness. Consistent with these findings, elevated MBNL1 expression in human breast tumors is associated with reduced metastatic relapse likelihood. Our findings delineate a post-transcriptional network that governs breast cancer metastasis through RNA-binding protein-mediated transcript stabilization.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al ARN/metabolismo , Animales , Neoplasias de la Mama/genética , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Unión Proteica , Estabilidad Proteica , Proteínas de Unión al ARN/genética , Recurrencia , Migración Transendotelial y Transepitelial/genética
16.
Nature ; 519(7544): 482-5, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25799998

RESUMEN

The first step in the biogenesis of microRNAs is the processing of primary microRNAs (pri-miRNAs) by the microprocessor complex, composed of the RNA-binding protein DGCR8 and the type III RNase DROSHA. This initial event requires recognition of the junction between the stem and the flanking single-stranded RNA of the pri-miRNA hairpin by DGCR8 followed by recruitment of DROSHA, which cleaves the RNA duplex to yield the pre-miRNA product. While the mechanisms underlying pri-miRNA processing have been determined, the mechanism by which DGCR8 recognizes and binds pri-miRNAs, as opposed to other secondary structures present in transcripts, is not understood. Here we find in mammalian cells that methyltransferase-like 3 (METTL3) methylates pri-miRNAs, marking them for recognition and processing by DGCR8. Consistent with this, METTL3 depletion reduced the binding of DGCR8 to pri-miRNAs and resulted in the global reduction of mature miRNAs and concomitant accumulation of unprocessed pri-miRNAs. In vitro processing reactions confirmed the sufficiency of the N(6)-methyladenosine (m(6)A) mark in promoting pri-miRNA processing. Finally, gain-of-function experiments revealed that METTL3 is sufficient to enhance miRNA maturation in a global and non-cell-type-specific manner. Our findings reveal that the m(6)A mark acts as a key post-transcriptional modification that promotes the initiation of miRNA biogenesis.


Asunto(s)
Adenosina/análogos & derivados , MicroARNs/química , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Adenosina/metabolismo , Secuencia de Bases , Línea Celular , Regulación de la Expresión Génica , Humanos , Metilación , Metiltransferasas/deficiencia , Metiltransferasas/metabolismo , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Proteínas de Unión al ARN/metabolismo , Especificidad por Sustrato
17.
EMBO J ; 35(1): 62-76, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26620550

RESUMEN

Altered abundance of phosphatidyl inositides (PIs) is a feature of cancer. Various PIs mark the identity of diverse membranes in normal and malignant cells. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) resides predominantly in the plasma membrane, where it regulates cellular processes by recruiting, activating, or inhibiting proteins at the plasma membrane. We find that PTPRN2 and PLCß1 enzymatically reduce plasma membrane PI(4,5)P2 levels in metastatic breast cancer cells through two independent mechanisms. These genes are upregulated in highly metastatic breast cancer cells, and their increased expression associates with human metastatic relapse. Reduction in plasma membrane PI(4,5)P2 abundance by these enzymes releases the PI(4,5)P2-binding protein cofilin from its inactive membrane-associated state into the cytoplasm where it mediates actin turnover dynamics, thereby enhancing cellular migration and metastatic capacity. Our findings reveal an enzymatic network that regulates metastatic cell migration through lipid-dependent sequestration of an actin-remodeling factor.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipasa C beta/metabolismo , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Animales , Neoplasias de la Mama , Línea Celular Tumoral , Humanos , Ratones SCID
18.
Nature ; 513(7517): 256-60, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25043050

RESUMEN

Aberrant regulation of RNA stability has an important role in many disease states. Deregulated post-transcriptional modulation, such as that governed by microRNAs targeting linear sequence elements in messenger RNAs, has been implicated in the progression of many cancer types. A defining feature of RNA is its ability to fold into structures. However, the roles of structural mRNA elements in cancer progression remain unexplored. Here we performed an unbiased search for post-transcriptional modulators of mRNA stability in breast cancer by conducting whole-genome transcript stability measurements in poorly and highly metastatic isogenic human breast cancer lines. Using a computational framework that searches RNA sequence and structure space, we discovered a family of GC-rich structural cis-regulatory RNA elements, termed sRSEs for structural RNA stability elements, which are significantly overrepresented in transcripts displaying reduced stability in highly metastatic cells. By integrating computational and biochemical approaches, we identified TARBP2, a double-stranded RNA-binding protein implicated in microRNA processing, as the trans factor that binds the sRSE family and similar structural elements--collectively termed TARBP2-binding structural elements (TBSEs)--in transcripts. TARBP2 is overexpressed in metastatic cells and metastatic human breast tumours and destabilizes transcripts containing TBSEs. Endogenous TARBP2 promotes metastatic cell invasion and colonization by destabilizing amyloid precursor protein (APP) and ZNF395 transcripts, two genes previously associated with Alzheimer's and Huntington's disease, respectively. We reveal these genes to be novel metastasis suppressor genes in breast cancer. The cleavage product of APP, extracellular amyloid-α peptide, directly suppresses invasion while ZNF395 transcriptionally represses a pro-metastatic gene expression program. The expression levels of TARBP2, APP and ZNF395 in human breast carcinomas support their experimentally uncovered roles in metastasis. Our findings establish a non-canonical and direct role for TARBP2 in mammalian gene expression regulation and reveal that regulated RNA destabilization through protein-mediated binding of mRNA structural elements can govern cancer progression.


Asunto(s)
Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Metástasis de la Neoplasia , Unión Proteica , Proteínas de Unión al ARN/genética , Factores de Transcripción/metabolismo
19.
Genes Dev ; 25(3): 226-31, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21289068

RESUMEN

Post-transcriptional regulators have emerged as robust effectors of metastasis and display deregulated expression through unknown mechanisms. Here, we reveal that the human microRNA-335 locus undergoes genetic deletion and epigenetic promoter hypermethylation in every metastatic derivative obtained from independent patients' malignant cell populations. Genetic deletion of miR-335 is a common event in human breast cancer, is enriched for in breast cancer metastases, and also correlates with ovarian cancer recurrence. We furthermore identify miR-335 as a robust inhibitor of tumor reinitiation. We thus implicate the miR-335 locus on 7q32.2 as the first selective metastasis suppressor and tumor initiation suppressor locus in human breast cancer.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Epigénesis Genética , MicroARNs/genética , MicroARNs/metabolismo , Metástasis de la Neoplasia/fisiopatología , Interferencia de ARN , Línea Celular Tumoral , Metilación de ADN , Femenino , Eliminación de Gen , Humanos , Metástasis de la Neoplasia/genética , Regiones Promotoras Genéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA