Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nat Metab ; 5(3): 495-515, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36941451

RESUMEN

Muscle degeneration is the most prevalent cause for frailty and dependency in inherited diseases and ageing. Elucidation of pathophysiological mechanisms, as well as effective treatments for muscle diseases, represents an important goal in improving human health. Here, we show that the lipid synthesis enzyme phosphatidylethanolamine cytidyltransferase (PCYT2/ECT) is critical to muscle health. Human deficiency in PCYT2 causes a severe disease with failure to thrive and progressive weakness. pcyt2-mutant zebrafish and muscle-specific Pcyt2-knockout mice recapitulate the participant phenotypes, with failure to thrive, progressive muscle weakness and accelerated ageing. Mechanistically, muscle Pcyt2 deficiency affects cellular bioenergetics and membrane lipid bilayer structure and stability. PCYT2 activity declines in ageing muscles of mice and humans, and adeno-associated virus-based delivery of PCYT2 ameliorates muscle weakness in Pcyt2-knockout and old mice, offering a therapy for individuals with a rare disease and muscle ageing. Thus, PCYT2 plays a fundamental and conserved role in vertebrate muscle health, linking PCYT2 and PCYT2-synthesized lipids to severe muscle dystrophy and ageing.


Asunto(s)
Insuficiencia de Crecimiento , ARN Nucleotidiltransferasas , Animales , Humanos , Ratones , Ratones Noqueados , Debilidad Muscular/genética , Músculos , ARN Nucleotidiltransferasas/química , ARN Nucleotidiltransferasas/genética , Pez Cebra
3.
Life Sci Alliance ; 6(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36725334

RESUMEN

Phagocytosis, the process by which cells engulf large particles, plays a vital role in driving tissue clearance and host defense. Its dysregulation is connected to autoimmunity, toxic accumulation of proteins, and increased risks for infections. Despite its importance, we lack full understanding of all molecular components involved in the process. To create a functional map in human cells, we performed a genome-wide CRISPRko FACS screen that identified 716 genes. Mapping those hits to a comprehensive protein-protein interaction network annotated for functional cellular processes allowed retrieval of protein complexes identified multiple times and detection of missing phagocytosis regulators. In addition to known components, such as the Arp2/3 complex, the vacuolar-ATPase-Rag machinery, and the Wave-2 complex, we identified and validated new phagocytosis-relevant functions, including the oligosaccharyltransferase complex (MAGT1/SLC58A1, DDOST, STT3B, and RPN2) and the hypusine pathway (eIF5A, DHPS, and DOHH). Overall, our phagocytosis network comprises elements of cargo uptake, shuffling, and biotransformation through the cell, providing a resource for the identification of potential novel drivers for diseases of the endo-lysosomal system. Our approach of integrating protein-protein interaction offers a broadly applicable way to functionally interpret genome-wide screens.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Hexosiltransferasas , Humanos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Proteínas , Fagocitosis/genética , Hexosiltransferasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo
4.
Life Sci Alliance ; 5(11)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36114003

RESUMEN

Solute carrier (SLC) transporters control fluxes of nutrients and metabolites across membranes and thereby represent a critical interface between the microenvironment and cellular and subcellular metabolism. Because of substantial functional overlap, the interplay and relative contributions of SLCs in response to environmental stresses remain poorly elucidated. To infer functional relationships between SLCs and metabolites, we developed a strategy to identify SLCs able to sustain cell viability and proliferation under growth-limiting concentrations of essential nutrients. One-by-one depletion of 13 amino acids required for cell proliferation enabled gain-of-function genetic screens using a SLC-focused CRISPR/Cas9-based transcriptional activation approach to uncover transporters relieving cells from growth-limiting metabolic bottlenecks. Among the transporters identified, we characterized the cationic amino acid transporter SLC7A3 as a gene that, when up-regulated, overcame low availability of arginine and lysine by increasing their uptake, whereas SLC7A5 was able to sustain cellular fitness upon deprivation of several neutral amino acids. Moreover, we identified metabolic compensation mediated by the glutamate/aspartate transporters SLC1A2 and SLC1A3 under glutamine-limiting conditions. Overall, this gain-of-function approach using human cells uncovered functional transporter-nutrient relationships and revealed that transport activity up-regulation may be sufficient to overcome environmental metabolic restrictions.


Asunto(s)
Proteínas de Transporte de Membrana , Nutrientes , Sistemas de Transporte de Aminoácidos Básicos/genética , Aminoácidos/metabolismo , Arginina/metabolismo , Ácido Aspártico/metabolismo , Mutación con Ganancia de Función , Glutamatos/metabolismo , Glutamina/metabolismo , Humanos , Transportador de Aminoácidos Neutros Grandes 1 , Lisina/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Nutrientes/metabolismo
5.
Nat Metab ; 3(5): 651-664, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33972798

RESUMEN

Metabolism negotiates cell-endogenous requirements of energy, nutrients and building blocks with the immediate environment to enable various processes, including growth and differentiation. While there is an increasing number of examples of crosstalk between metabolism and chromatin, few involve uptake of exogenous metabolites. Solute carriers (SLCs) represent the largest group of transporters in the human genome and are responsible for the transport of a wide variety of substrates, including nutrients and metabolites. We aimed to investigate the possible involvement of SLC-mediated solutes uptake and cellular metabolism in regulating cellular epigenetic states. Here, we perform a CRISPR-Cas9 transporter-focused genetic screen and a metabolic compound library screen for the regulation of BRD4-dependent chromatin states in human myeloid leukaemia cells. Intersection of the two orthogonal approaches reveal that loss of transporters involved with purine transport or inhibition of de novo purine synthesis lead to dysfunction of BRD4-dependent transcriptional regulation. Through mechanistic characterization of the metabolic circuitry, we elucidate the convergence of SLC-mediated purine uptake and de novo purine synthesis on BRD4-chromatin occupancy. Moreover, adenine-related metabolite supplementation effectively restores BRD4 functionality on purine impairment. Our study highlights the specific role of purine/adenine metabolism in modulating BRD4-dependent epigenetic states.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas de Transporte de Nucleósidos/metabolismo , Purinas/metabolismo , Proteínas Transportadoras de Solutos/metabolismo , Factores de Transcripción/metabolismo , Adenina/metabolismo , Vías Biosintéticas , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular , Cromatina/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de Transporte de Membrana , Modelos Biológicos , Proteínas Transportadoras de Solutos/genética , Factores de Transcripción/antagonistas & inhibidores , Transcripción Genética
6.
Nat Commun ; 11(1): 6145, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262325

RESUMEN

About a thousand genes in the human genome encode for membrane transporters. Among these, several solute carrier proteins (SLCs), representing the largest group of transporters, are still orphan and lack functional characterization. We reasoned that assessing genetic interactions among SLCs may be an efficient way to obtain functional information allowing their deorphanization. Here we describe a network of strong genetic interactions indicating a contribution to mitochondrial respiration and redox metabolism for SLC25A51/MCART1, an uncharacterized member of the SLC25 family of transporters. Through a combination of metabolomics, genomics and genetics approaches, we demonstrate a role for SLC25A51 as enabler of mitochondrial import of NAD, showcasing the potential of genetic interaction-driven functional gene deorphanization.


Asunto(s)
Epistasis Genética , Mitocondrias/metabolismo , NAD/metabolismo , Proteína Desacopladora 1/metabolismo , Transporte Biológico , Humanos , Mitocondrias/genética , Oxidación-Reducción , Proteína Desacopladora 1/genética
7.
Front Immunol ; 11: 1398, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733464

RESUMEN

The bioavailability of the major pro-inflammatory cytokines IL-1α and IL-1ß is tightly controlled by transcription and post-translational processing to prevent hyperinflammation. The role of mRNA decay in maintenance of physiological IL-1 amounts remained unknown. Here we show that the down-regulation of Il1a and Il1b mRNA by the mRNA-destabilizing protein TTP (gene Zfp36) is required for immune homeostasis. The TTP deficiency syndrome, a multi organ inflammation in TTP-/- mice, was significantly ameliorated upon deletion of the IL-1 receptor. Il1a and Il1b played non-redundant roles in triggering the pathological IL-1 signaling in TTP-/- mice. Accordingly, tissues from TTP-/- animals contained increased amounts of Il1b mRNA. Unexpectedly, TTP destabilized Il1b mRNA in cell type-specific ways as evident from RNA-Seq and mRNA stability assays. These results demonstrate that TTP-driven mRNA destabilization depends on the cellular context. Moreover, such context-defined mRNA decay is essential for keeping steady state IL-1 levels in the physiological range.


Asunto(s)
Regulación de la Expresión Génica , Homeostasis , Inmunidad/genética , Interleucina-1/genética , Tristetraprolina/metabolismo , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Inflamación/diagnóstico , Inflamación/etiología , Inflamación/metabolismo , Interleucina-1/metabolismo , Interleucina-1alfa/genética , Interleucina-1beta/genética , Ratones , Ratones Noqueados , Estabilidad del ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Índice de Severidad de la Enfermedad
8.
Mol Syst Biol ; 16(7): e9652, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32697042

RESUMEN

Solute carriers (SLCs) are the largest family of transmembrane transporters in the human genome with more than 400 members. Despite the fact that SLCs mediate critical biological functions and several are important pharmacological targets, a large proportion of them is poorly characterized and present no assigned substrate. A major limitation to systems-level de-orphanization campaigns is the absence of a structured, language-controlled chemical annotation. Here we describe a thorough manual annotation of SLCs based on literature. The annotation of substrates, transport mechanism, coupled ions, and subcellular localization for 446 human SLCs confirmed that ~30% of these were still functionally orphan and lacked known substrates. Application of a substrate-based ontology to transcriptomic datasets identified SLC-specific responses to external perturbations, while a machine-learning approach based on the annotation allowed us to identify potential substrates for several orphan SLCs. The annotation is available at https://opendata.cemm.at/gsflab/slcontology/. Given the increasing availability of large biological datasets and the growing interest in transporters, we expect that the effort presented here will be critical to provide novel insights into the functions of SLCs.


Asunto(s)
Transporte Biológico Activo/genética , Biología Computacional/métodos , Proteínas de Transporte de Membrana/metabolismo , Aminoácidos/metabolismo , Aminoácidos/farmacología , Ontologías Biológicas , Línea Celular , Perfilación de la Expresión Génica , Genoma Humano , Humanos , Aprendizaje Automático
9.
Nature ; 581(7808): 316-322, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32433612

RESUMEN

Toll-like receptors (TLRs) have a crucial role in the recognition of pathogens and initiation of immune responses1-3. Here we show that a previously uncharacterized protein encoded by CXorf21-a gene that is associated with systemic lupus erythematosus4,5-interacts with the endolysosomal transporter SLC15A4, an essential but poorly understood component of the endolysosomal TLR machinery also linked to autoimmune disease4,6-9. Loss of this type-I-interferon-inducible protein, which we refer to as 'TLR adaptor interacting with SLC15A4 on the lysosome' (TASL), abrogated responses to endolysosomal TLR agonists in both primary and transformed human immune cells. Deletion of SLC15A4 or TASL specifically impaired the activation of the IRF pathway without affecting NF-κB and MAPK signalling, which indicates that ligand recognition and TLR engagement in the endolysosome occurred normally. Extensive mutagenesis of TASL demonstrated that its localization and function relies on the interaction with SLC15A4. TASL contains a conserved pLxIS motif (in which p denotes a hydrophilic residue and x denotes any residue) that mediates the recruitment and activation of IRF5. This finding shows that TASL is an innate immune adaptor for TLR7, TLR8 and TLR9 signalling, revealing a clear mechanistic analogy with the IRF3 adaptors STING, MAVS and TRIF10,11. The identification of TASL as the component that links endolysosomal TLRs to the IRF5 transcription factor via SLC15A4 provides a mechanistic explanation for the involvement of these proteins in systemic lupus erythematosus12-14.


Asunto(s)
Factores Reguladores del Interferón/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Receptor Toll-Like 9/metabolismo , Secuencias de Aminoácidos , Animales , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Lupus Eritematoso Sistémico/metabolismo , Masculino , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Unión Proteica , Transducción de Señal
11.
Nat Chem Biol ; 16(4): 469-478, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32152546

RESUMEN

Solute carriers (SLCs) are the largest family of transmembrane transporters in humans and are major determinants of cellular metabolism. Several SLCs have been shown to be required for the uptake of chemical compounds into cellular systems, but systematic surveys of transporter-drug relationships in human cells are currently lacking. We performed a series of genetic screens in a haploid human cell line against 60 cytotoxic compounds representative of the chemical space populated by approved drugs. By using an SLC-focused CRISPR-Cas9 library, we identified transporters whose absence induced resistance to the drugs tested. This included dependencies involving the transporters SLC11A2/SLC16A1 for artemisinin derivatives and SLC35A2/SLC38A5 for cisplatin. The functional dependence on SLCs observed for a significant proportion of the screened compounds suggests a widespread role for SLCs in the uptake and cellular activity of cytotoxic drugs and provides an experimentally validated set of SLC-drug associations for a number of clinically relevant compounds.


Asunto(s)
Resistencia a Medicamentos/genética , Proteínas Transportadoras de Solutos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Antineoplásicos , Fenómenos Bioquímicos , Transporte Biológico/genética , Transporte Biológico/fisiología , Sistemas CRISPR-Cas , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Resistencia a Medicamentos/fisiología , Pruebas Genéticas , Humanos , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Transporte de Proteínas/fisiología , Proteínas Transportadoras de Solutos/fisiología , Simportadores/genética , Simportadores/metabolismo
12.
Mol Cell ; 76(3): 485-499.e8, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31495563

RESUMEN

Transcriptional responses to external stimuli remain poorly understood. Using global nuclear run-on followed by sequencing (GRO-seq) and precision nuclear run-on sequencing (PRO-seq), we show that CDK8 kinase activity promotes RNA polymerase II pause release in response to interferon-γ (IFN-γ), a universal cytokine involved in immunity and tumor surveillance. The Mediator kinase module contains CDK8 or CDK19, which are presumed to be functionally redundant. We implemented cortistatin A, chemical genetics, transcriptomics, and other methods to decouple their function while assessing enzymatic versus structural roles. Unexpectedly, CDK8 and CDK19 regulated different gene sets via distinct mechanisms. CDK8-dependent regulation required its kinase activity, whereas CDK19 governed IFN-γ responses through its scaffolding function (i.e., it was kinase independent). Accordingly, CDK8, not CDK19, phosphorylates the STAT1 transcription factor (TF) during IFN-γ stimulation, and CDK8 kinase inhibition blocked activation of JAK-STAT pathway TFs. Cytokines such as IFN-γ rapidly mobilize TFs to "reprogram" cellular transcription; our results implicate CDK8 and CDK19 as essential for this transcriptional reprogramming.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Fibroblastos/efectos de los fármacos , Interferón gamma/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Quinasa 8 Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/genética , Fibroblastos/enzimología , Fibroblastos/virología , Células HCT116 , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , ARN Polimerasa II/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Vesiculovirus/patogenicidad
13.
Science ; 362(6419): 1171-1177, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30442766

RESUMEN

In genetic screens aimed at understanding drug resistance mechanisms in chronic myeloid leukemia cells, inactivation of the cullin 3 adapter protein-encoding leucine zipper-like transcription regulator 1 (LZTR1) gene led to enhanced mitogen-activated protein kinase (MAPK) pathway activity and reduced sensitivity to tyrosine kinase inhibitors. Knockdown of the Drosophila LZTR1 ortholog CG3711 resulted in a Ras-dependent gain-of-function phenotype. Endogenous human LZTR1 associates with the main RAS isoforms. Inactivation of LZTR1 led to decreased ubiquitination and enhanced plasma membrane localization of endogenous KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog). We propose that LZTR1 acts as a conserved regulator of RAS ubiquitination and MAPK pathway activation. Because LZTR1 disease mutations failed to revert loss-of-function phenotypes, our findings provide a molecular rationale for LZTR1 involvement in a variety of inherited and acquired human disorders.


Asunto(s)
Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/fisiología , Ubiquitinación , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Drosophila melanogaster , Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Mutación con Ganancia de Función , Técnicas de Silenciamiento del Gen , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/epidemiología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Mutación con Pérdida de Función , Sistema de Señalización de MAP Quinasas/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridazinas/farmacología , Piridazinas/uso terapéutico , Transducción de Señal , Factores de Transcripción/genética , Ubiquitinación/genética
14.
Cell Host Microbe ; 23(6): 766-774.e5, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29779931

RESUMEN

Macrophages represent the first line of immune defense against pathogens, and phagosome acidification is a necessary step in pathogen clearance. Here, we identified the bicarbonate transporter SLC4A7, which is strongly induced upon macrophage differentiation, as critical for phagosome acidification. Loss of SLC4A7 reduced acidification of phagocytosed beads or bacteria and impaired the intracellular microbicidal capacity in human macrophage cell lines. The phenotype was rescued by wild-type SLC4A7, but not by SLC4A7 mutants, affecting transport capacity or cell surface localization. Loss of SLC4A7 resulted in increased cytoplasmic acidification during phagocytosis, suggesting that SLC4A7-mediated, bicarbonate-driven maintenance of cytoplasmic pH is necessary for phagosome acidification. Altogether, we identify SLC4A7 and bicarbonate-driven cytoplasmic pH homeostasis as an important element of phagocytosis and the associated microbicidal functions in macrophages.


Asunto(s)
Bicarbonatos/metabolismo , Macrófagos/metabolismo , Fagosomas/metabolismo , Simportadores de Sodio-Bicarbonato/fisiología , Sistemas CRISPR-Cas , Proteínas de Transporte de Catión/metabolismo , Citoplasma/metabolismo , Técnicas de Inactivación de Genes , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Fagocitosis , Simportadores de Sodio-Bicarbonato/genética , Células THP-1 , Transcriptoma , Células U937
15.
Mol Cell ; 67(1): 30-43.e6, 2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28648779

RESUMEN

In search for RNA signals that modulate transcription via direct interaction with RNA polymerase (RNAP), we deep sequenced an E. coli genomic library enriched for RNAP-binding RNAs. Many natural RNAP-binding aptamers, termed RAPs, were mapped to the genome. Over 60% of E. coli genes carry RAPs in their mRNA. Combining in vitro and in vivo approaches, we characterized a subset of inhibitory RAPs (iRAPs) that promote Rho-dependent transcription termination. A representative iRAP within the coding region of the essential gene, nadD, greatly reduces its transcriptional output in stationary phase and under oxidative stress, demonstrating that iRAPs control gene expression in response to changing environment. The mechanism of iRAPs involves active uncoupling of transcription and translation, making nascent RNA accessible to Rho. iRAPs encoded in the antisense strand also promote gene expression by reducing transcriptional interference. In essence, our work uncovers a broad class of cis-acting RNA signals that globally control bacterial transcription.


Asunto(s)
Aptámeros de Nucleótidos/genética , Proteínas Bacterianas/genética , ARN Polimerasas Dirigidas por ADN/genética , Escherichia coli/genética , Técnica SELEX de Producción de Aptámeros , Terminación de la Transcripción Genética , Aptámeros de Nucleótidos/metabolismo , Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/enzimología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Nicotinamida-Nucleótido Adenililtransferasa/genética , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Sistemas de Lectura Abierta , Ribosomas/metabolismo , Factores de Tiempo
16.
J Clin Invest ; 127(6): 2051-2065, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28504646

RESUMEN

Protective responses against pathogens require a rapid mobilization of resting neutrophils and the timely removal of activated ones. Neutrophils are exceptionally short-lived leukocytes, yet it remains unclear whether the lifespan of pathogen-engaged neutrophils is regulated differently from that in the circulating steady-state pool. Here, we have found that under homeostatic conditions, the mRNA-destabilizing protein tristetraprolin (TTP) regulates apoptosis and the numbers of activated infiltrating murine neutrophils but not neutrophil cellularity. Activated TTP-deficient neutrophils exhibited decreased apoptosis and enhanced accumulation at the infection site. In the context of myeloid-specific deletion of Ttp, the potentiation of neutrophil deployment protected mice against lethal soft tissue infection with Streptococcus pyogenes and prevented bacterial dissemination. Neutrophil transcriptome analysis revealed that decreased apoptosis of TTP-deficient neutrophils was specifically associated with elevated expression of myeloid cell leukemia 1 (Mcl1) but not other antiapoptotic B cell leukemia/lymphoma 2 (Bcl2) family members. Higher Mcl1 expression resulted from stabilization of Mcl1 mRNA in the absence of TTP. The low apoptosis rate of infiltrating TTP-deficient neutrophils was comparable to that of transgenic Mcl1-overexpressing neutrophils. Our study demonstrates that posttranscriptional gene regulation by TTP schedules the termination of the antimicrobial engagement of neutrophils. The balancing role of TTP comes at the cost of an increased risk of bacterial infections.


Asunto(s)
Apoptosis/inmunología , Infecciones Estreptocócicas/metabolismo , Tristetraprolina/fisiología , Animales , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Unión Proteica , Estabilidad del ARN , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Transcriptoma/inmunología
17.
Cancer Res ; 77(9): 2424-2438, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28428272

RESUMEN

HuR is an RNA-binding protein implicated in immune homeostasis and various cancers, including colorectal cancer. HuR binding to AU-rich elements within the 3' untranslated region of mRNAs encoding oncogenes, growth factors, and various cytokines leads message stability and translation. In this study, we evaluated HuR as a small-molecule target for preventing colorectal cancer in high-risk groups such as those with familial adenomatosis polyposis (FAP) or inflammatory bowel disease (IBD). In human specimens, levels of cytoplasmic HuR were increased in colonic epithelial cells from patients with IBD, IBD-cancer, FAP-adenoma, and colorectal cancer, but not in patients with IBD-dysplasia. Intraperitoneal injection of the HuR small-molecule inhibitor MS-444 in AOM/DSS mice, a model of IBD and inflammatory colon cancer, augmented DSS-induced weight loss and increased tumor multiplicity, size, and invasiveness. MS-444 treatment also abrogated tumor cell apoptosis and depleted tumor-associated eosinophils, accompanied by a decrease in IL18 and eotaxin-1. In contrast, HuR inhibition in APCMin mice, a model of FAP and colon cancer, diminished the number of small intestinal tumors generated. In this setting, fecal microbiota, evaluated by 16S rRNA gene amplicon sequencing, shifted to a state of reduced bacterial diversity, with an increased representation of Prevotella, Akkermansia, and Lachnospiraceae Taken together, our results indicate that HuR activation is an early event in FAP-adenoma but is not present in IBD-dysplasia. Furthermore, our results offer a preclinical proof of concept for HuR inhibition as an effective means of FAP chemoprevention, with caution advised in the setting of IBD. Cancer Res; 77(9); 2424-38. ©2017 AACR.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Neoplasias Colorrectales/genética , Proteína 1 Similar a ELAV/genética , Enfermedades Inflamatorias del Intestino/genética , Poliposis Adenomatosa del Colon/microbiología , Poliposis Adenomatosa del Colon/patología , Animales , Apoptosis/efectos de los fármacos , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Quimiocina CCL11/genética , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Proteína 1 Similar a ELAV/antagonistas & inhibidores , Heces/microbiología , Furanos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Células HCT116 , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Interleucina-18/genética , Ratones , Naftoles/administración & dosificación , Células RAW 264.7
18.
Cytokine ; 89: 21-26, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26586165

RESUMEN

Expression of cytokines and chemokines is regulated at multiple steps during the transfer of the genetic information from DNA sequence to the functional protein. The multilayered control of cytokine expression reflects the need of the immune system to precisely and rapidly adjust the magnitude and duration of immune responses to external cues. Common features of the regulation of cytokine expression are temporal and highly dynamic changes in cytokine mRNA stability. Failures in the timing and extent of mRNA decay can result in disease. Recent advances in transcriptome-wide approaches began to shed light into the complex network of cis-acting sequence elements and trans-acting factors controlling mRNA stability. These approaches led to the discovery of novel unexpected paradigms but they also revealed new questions. This review will discuss the control of cytokine mRNA stability both in the context of high content approaches as well as focused mechanistic studies and animal models. The article highlights the need for systems biology approaches as important means to understand how cytokine mRNA decay helps maintain the immune and tissue homeostasis, and to explore options for therapeutical exploitation of mRNA stability regulation.


Asunto(s)
Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Estabilidad del ARN/inmunología , ARN Mensajero/inmunología , Transcriptoma/inmunología , Animales , Humanos
19.
Mol Syst Biol ; 12(5): 868, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27178967

RESUMEN

Precise regulation of mRNA decay is fundamental for robust yet not exaggerated inflammatory responses to pathogens. However, a global model integrating regulation and functional consequences of inflammation-associated mRNA decay remains to be established. Using time-resolved high-resolution RNA binding analysis of the mRNA-destabilizing protein tristetraprolin (TTP), an inflammation-limiting factor, we qualitatively and quantitatively characterize TTP binding positions in the transcriptome of immunostimulated macrophages. We identify pervasive destabilizing and non-destabilizing TTP binding, including a robust intronic binding, showing that TTP binding is not sufficient for mRNA destabilization. A low degree of flanking RNA structuredness distinguishes occupied from silent binding motifs. By functionally relating TTP binding sites to mRNA stability and levels, we identify a TTP-controlled switch for the transition from inflammatory into the resolution phase of the macrophage immune response. Mapping of binding positions of the mRNA-stabilizing protein HuR reveals little target and functional overlap with TTP, implying a limited co-regulation of inflammatory mRNA decay by these proteins. Our study establishes a functionally annotated and navigable transcriptome-wide atlas (http://ttp-atlas.univie.ac.at) of cis-acting elements controlling mRNA decay in inflammation.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/inmunología , ARN Mensajero/química , Tristetraprolina/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Células HEK293 , Humanos , Macrófagos/efectos de los fármacos , Ratones , Estabilidad del ARN , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
20.
Nucleic Acids Res ; 44(D1): D90-5, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26602692

RESUMEN

AREsite2 represents an update for AREsite, an on-line resource for the investigation of AU-rich elements (ARE) in human and mouse mRNA 3'UTR sequences. The new updated and enhanced version allows detailed investigation of AU, GU and U-rich elements (ARE, GRE, URE) in the transcriptome of Homo sapiens, Mus musculus, Danio rerio, Caenorhabditis elegans and Drosophila melanogaster. It contains information on genomic location, genic context, RNA secondary structure context and conservation of annotated motifs. Improvements include annotation of motifs not only in 3'UTRs but in the whole gene body including introns, additional genomes, and locally stable secondary structures from genome wide scans. Furthermore, we include data from CLIP-Seq experiments in order to highlight motifs with validated protein interaction. Additionally, we provide a REST interface for experienced users to interact with the database in a semi-automated manner. The database is publicly available at: http://rna.tbi.univie.ac.at/AREsite.


Asunto(s)
Regiones no Traducidas 3' , Bases de Datos de Ácidos Nucleicos , ARN/química , Animales , Genómica , Humanos , Ratones , Anotación de Secuencia Molecular , Conformación de Ácido Nucleico , Motivos de Nucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...