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

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 84(6): 991-994, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38518748

RESUMO

We spoke with authors co-first authors Jayoung Ku, Keonyong Lee, and lead author Yoosik Kim about their paper "Alternative polyadenylation determines the functional landscape of inverted Alu repeats" (this issue of Molecular Cell), finding renewed energy from attending scientific meetings, and the open questions they are most interested in investigating next.

2.
Mol Cell ; 84(15): 2935-2948.e7, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39019044

RESUMO

Mitochondria are essential regulators of innate immunity. They generate long mitochondrial double-stranded RNAs (mt-dsRNAs) and release them into the cytosol to trigger an immune response under pathological stress conditions. Yet the regulation of these self-immunogenic RNAs remains largely unknown. Here, we employ CRISPR screening on mitochondrial RNA (mtRNA)-binding proteins and identify NOP2/Sun RNA methyltransferase 4 (NSUN4) as a key regulator of mt-dsRNA expression in human cells. We find that NSUN4 induces 5-methylcytosine (m5C) modification on mtRNAs, especially on the termini of light-strand long noncoding RNAs. These m5C-modified RNAs are recognized by complement C1q-binding protein (C1QBP), which recruits polyribonucleotide nucleotidyltransferase to facilitate RNA turnover. Suppression of NSUN4 or C1QBP results in increased mt-dsRNA expression, while C1QBP deficiency also leads to increased cytosolic mt-dsRNAs and subsequent immune activation. Collectively, our study unveils the mechanism underlying the selective degradation of light-strand mtRNAs and establishes a molecular mark for mtRNA decay and cytosolic release.


Assuntos
5-Metilcitosina , Citosol , Mitocôndrias , Estabilidade de RNA , RNA de Cadeia Dupla , RNA Mitocondrial , Humanos , Citosol/metabolismo , 5-Metilcitosina/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/genética , RNA de Cadeia Dupla/metabolismo , RNA de Cadeia Dupla/genética , RNA Mitocondrial/genética , RNA Mitocondrial/metabolismo , Células HEK293 , Células HeLa , Metiltransferases/metabolismo , Metiltransferases/genética , Imunidade Inata , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Sistemas CRISPR-Cas
3.
Mol Cell ; 84(6): 1062-1077.e9, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38309276

RESUMO

Inverted Alu repeats (IRAlus) are abundantly found in the transcriptome, especially in introns and 3' untranslated regions (UTRs). Yet, the biological significance of IRAlus embedded in 3' UTRs remains largely unknown. Here, we find that 3' UTR IRAlus silences genes involved in essential signaling pathways. We utilize J2 antibody to directly capture and map the double-stranded RNA structure of 3' UTR IRAlus in the transcriptome. Bioinformatic analysis reveals alternative polyadenylation as a major axis of IRAlus-mediated gene regulation. Notably, the expression of mouse double minute 2 (MDM2), an inhibitor of p53, is upregulated by the exclusion of IRAlus during UTR shortening, which is exploited to silence p53 during tumorigenesis. Moreover, the transcriptome-wide UTR lengthening in neural progenitor cells results in the global downregulation of genes associated with neurodegenerative diseases, including amyotrophic lateral sclerosis, via IRAlus inclusion. Our study establishes the functional landscape of 3' UTR IRAlus and its role in human pathophysiology.


Assuntos
Poliadenilação , Proteína Supressora de Tumor p53 , Humanos , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Regiões 3' não Traduzidas/genética , Regulação da Expressão Gênica , Íntrons
4.
Cell ; 151(4): 765-777, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23102813

RESUMO

LIN28 plays a critical role in developmental transition, glucose metabolism, and tumorigenesis. At the molecular level, LIN28 is known to repress maturation of let-7 microRNAs and enhance translation of certain mRNAs. In this study, we obtain a genome-wide view of the molecular function of LIN28A in mouse embryonic stem cells by carrying out RNA crosslinking-immunoprecipitation-sequencing (CLIP-seq) and ribosome footprinting. We find that, in addition to let-7 precursors, LIN28A binds to a large number of spliced mRNAs. LIN28A recognizes AAGNNG, AAGNG, and less frequently UGUG, which are located in the terminal loop of a small hairpin. LIN28A is localized to the periendoplasmic reticulum (ER) area and inhibits translation of mRNAs that are destined for the ER, reducing the synthesis of transmembrane proteins, ER or Golgi lumen proteins, and secretory proteins. Our study suggests a selective regulatory mechanism for ER-associated translation and reveals an unexpected role of LIN28A as a global suppressor of genes in the secretory pathway.


Assuntos
Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Células-Tronco Embrionárias/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Imunoprecipitação/métodos , Camundongos , MicroRNAs/metabolismo , Ribossomos/metabolismo , Via Secretória , Análise de Sequência de RNA
5.
Mol Cell ; 71(6): 1051-1063.e6, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30174290

RESUMO

Protein kinase RNA-activated (PKR) induces immune response by sensing viral double-stranded RNAs (dsRNAs). However, growing evidence suggests that PKR can also be activated by endogenously expressed dsRNAs. Here, we capture these dsRNAs by formaldehyde-mediated crosslinking and immunoprecipitation sequencing and find that various noncoding RNAs interact with PKR. Surprisingly, the majority of the PKR-interacting RNA repertoire is occupied by mitochondrial RNAs (mtRNAs). MtRNAs can form intermolecular dsRNAs owing to bidirectional transcription of the mitochondrial genome and regulate PKR and eIF2α phosphorylation to control cell signaling and translation. Moreover, PKR activation by mtRNAs is counteracted by PKR phosphatases, disruption of which causes apoptosis from PKR overactivation even in uninfected cells. Our work unveils dynamic regulation of PKR even without infection and establishes PKR as a sensor for nuclear and mitochondrial signaling cues in regulating cellular metabolism.


Assuntos
eIF-2 Quinase/metabolismo , eIF-2 Quinase/fisiologia , Linhagem Celular , Núcleo Celular , Ativação Enzimática , Fator de Iniciação 2 em Eucariotos/metabolismo , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação/métodos , Mitocôndrias/genética , Fosforilação , RNA de Cadeia Dupla/genética , RNA Mitocondrial/genética , RNA Mitocondrial/fisiologia , RNA não Traduzido/genética , RNA não Traduzido/fisiologia , Transdução de Sinais , eIF-2 Quinase/imunologia
6.
Mol Cell ; 62(3): 462-471, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153541

RESUMO

Poly(A) tails are critical for mRNA stability and translation. However, recent studies have challenged this view, showing that poly(A) tail length and translation efficiency are decoupled in non-embryonic cells. Using TAIL-seq and ribosome profiling, we investigate poly(A) tail dynamics and translational control in the somatic cell cycle. We find dramatic changes in poly(A) tail lengths of cell-cycle regulatory genes like CDK1, TOP2A, and FBXO5, explaining their translational repression in M phase. We also find that poly(A) tail length is coupled to translation when the poly(A) tail is <20 nucleotides. However, as most genes have >20 nucleotide poly(A) tails, their translation is regulated mainly via poly(A) tail length-independent mechanisms during the cell cycle. Specifically, we find that terminal oligopyrimidine (TOP) tract-containing transcripts escape global translational suppression in M phase and are actively translated. Our quantitative and comprehensive data provide a revised view of translational control in the somatic cell cycle.


Assuntos
Mitose , Poli A/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Proteína Quinase CDC2 , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Quinases Ciclina-Dependentes/biossíntese , Quinases Ciclina-Dependentes/genética , DNA Topoisomerases Tipo II/biossíntese , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas F-Box/biossíntese , Proteínas F-Box/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Biblioteca Gênica , Células HeLa , Humanos , Pontos de Checagem da Fase M do Ciclo Celular , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/metabolismo , Poli A/genética , Poli dA-dT/genética , Poli dA-dT/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Estabilidade de RNA , RNA Mensageiro/genética , Ribossomos/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular , Fatores de Tempo
7.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33762305

RESUMO

DNA-methyltransferase inhibitors (DNMTis), such as azacitidine and decitabine, are used clinically to treat myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Decitabine activates the transcription of endogenous retroviruses (ERVs), which can induce immune response by acting as cellular double-stranded RNAs (dsRNAs). Yet, the posttranscriptional regulation of ERV dsRNAs remains uninvestigated. Here, we find that the viral mimicry and subsequent cell death in response to decitabine require the dsRNA-binding protein Staufen1 (Stau1). We show that Stau1 directly binds to ERV RNAs and stabilizes them in a genome-wide manner. Furthermore, Stau1-mediated stabilization requires a long noncoding RNA TINCR, which enhances the interaction between Stau1 and ERV RNAs. Analysis of a clinical patient cohort reveals that MDS and AML patients with lower Stau1 and TINCR expressions exhibit inferior treatment outcomes to DNMTi therapy. Overall, our study reveals the posttranscriptional regulatory mechanism of ERVs and identifies the Stau1-TINCR complex as a potential target for predicting the efficacy of DNMTis and other drugs that rely on dsRNAs.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Proteínas do Citoesqueleto/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Estudos de Coortes , Proteínas do Citoesqueleto/genética , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/imunologia , Decitabina/farmacologia , Decitabina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Retrovirus Endógenos/genética , Feminino , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Células HCT116 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/mortalidade , Intervalo Livre de Progressão , Estabilidade de RNA/efeitos dos fármacos , Estabilidade de RNA/imunologia , RNA de Cadeia Dupla/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/genética , RNA-Seq
8.
Int J Mol Sci ; 24(8)2023 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-37108567

RESUMO

Human mitochondria contain a circular genome that encodes 13 subunits of the oxidative phosphorylation system. In addition to their role as powerhouses of the cells, mitochondria are also involved in innate immunity as the mitochondrial genome generates long double-stranded RNAs (dsRNAs) that can activate the dsRNA-sensing pattern recognition receptors. Recent evidence shows that these mitochondrial dsRNAs (mt-dsRNAs) are closely associated with the pathogenesis of human diseases that accompany inflammation and aberrant immune activation, such as Huntington's disease, osteoarthritis, and autoimmune Sjögren's syndrome. Yet, small chemicals that can protect cells from a mt-dsRNA-mediated immune response remain largely unexplored. Here, we investigate the potential of resveratrol (RES), a plant-derived polyphenol with antioxidant properties, on suppressing mt-dsRNA-mediated immune activation. We show that RES can revert the downstream response to immunogenic stressors that elevate mitochondrial RNA expressions, such as stimulation by exogenous dsRNAs or inhibition of ATP synthase. Through high-throughput sequencing, we find that RES can regulate mt-dsRNA expression, interferon response, and other cellular responses induced by these stressors. Notably, RES treatment fails to counter the effect of an endoplasmic reticulum stressor that does not affect the expression of mitochondrial RNAs. Overall, our study demonstrates the potential usage of RES to alleviate the mt-dsRNA-mediated immunogenic stress response.


Assuntos
Mitocôndrias , RNA de Cadeia Dupla , Humanos , Resveratrol/farmacologia , Resveratrol/metabolismo , RNA Mitocondrial/genética , Mitocôndrias/metabolismo , RNA de Cadeia Dupla/metabolismo , Imunidade Inata
9.
Proc Natl Acad Sci U S A ; 116(17): 8289-8294, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30948645

RESUMO

DNA-reactive compounds are harnessed for cancer chemotherapy. Their genotoxic effects are considered to be the main mechanism for the cytotoxicity to date. Because this mechanism preferentially affects actively proliferating cells, it is postulated that the cytotoxicity is specific to cancer cells. Nonetheless, they do harm normal quiescent cells, suggesting that there are other cytotoxic mechanisms to be uncovered. By employing doxorubicin as a representative DNA-reactive compound, we have discovered a cytotoxic mechanism that involves a cellular noncoding RNA (ncRNA) nc886 and protein kinase R (PKR) that is a proapoptotic protein. nc886 is transcribed by RNA polymerase III (Pol III), binds to PKR, and prevents it from aberrant activation in most normal cells. We have shown here that doxorubicin evicts Pol III from DNA and, thereby, shuts down nc886 transcription. Consequently, the instantaneous depletion of nc886 provokes PKR and leads to apoptosis. In a short-pulse treatment of doxorubicin, these events are the main cause of cytotoxicity preceding the DNA damage response in a 3D culture system as well as the monolayer cultures. By identifying nc886 as a molecular signal for PKR to sense doxorubicin, we have provided an explanation for the conundrum why DNA-damaging drugs can be cytotoxic to quiescent cells that have the competent nc886/PKR pathway.


Assuntos
Apoptose/efeitos dos fármacos , DNA/metabolismo , MicroRNAs/metabolismo , RNA não Traduzido , Linhagem Celular , Doxorrubicina/farmacologia , Humanos , MicroRNAs/genética , RNA Polimerase III/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo
10.
Genes Dev ; 28(12): 1310-22, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24939934

RESUMO

dsRNA-dependent protein kinase R (PKR) is a ubiquitously expressed enzyme well known for its roles in immune response. Upon binding to viral dsRNA, PKR undergoes autophosphorylation, and the phosphorylated PKR (pPKR) regulates translation and multiple signaling pathways in infected cells. Here, we found that PKR is activated in uninfected cells, specifically during mitosis, by binding to dsRNAs formed by inverted Alu repeats (IRAlus). While PKR and IRAlu-containing RNAs are segregated in the cytosol and nucleus of interphase cells, respectively, they interact during mitosis when nuclear structure is disrupted. Once phosphorylated, PKR suppresses global translation by phosphorylating the α subunit of eukaryotic initiation factor 2 (eIF2α). In addition, pPKR acts as an upstream kinase for c-Jun N-terminal kinase and regulates the levels of multiple mitotic factors such as cyclins A and B and Polo-like kinase 1 and phosphorylation of histone H3. Disruption of PKR activation via RNAi or expression of a transdominant-negative mutant leads to misregulation of the mitotic factors, delay in mitotic progression, and defects in cytokinesis. Our study unveils a novel function of PKR and endogenous dsRNAs as signaling molecules during the mitosis of uninfected cells.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Mitose/fisiologia , RNA de Cadeia Dupla/metabolismo , eIF-2 Quinase/metabolismo , Elementos Alu/fisiologia , Ciclo Celular/fisiologia , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica , Células HeLa , Humanos , Fosforilação , Ligação Proteica
11.
Bioessays ; 41(7): e1900023, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31099409

RESUMO

Many innate immune response proteins recognize foreign nucleic acids from invading pathogens to initiate antiviral signaling. These proteins mostly rely on structural characteristics of the nucleic acids rather than their specific sequences to distinguish self and nonself. One feature utilized by RNA sensors is the extended stretch of double-stranded RNA (dsRNA) base pairs. However, the criteria for recognizing nonself dsRNAs are rather lenient, and hairpin structure of self-RNAs can also trigger an immune response. Consequently, aberrant activation of RNA sensors has been reported in numerous human diseases. Yet, in most cases, the activating antigens remain unknown. Recent studies have developed sequencing techniques tailored to specifically capture dsRNAs and identified that various noncoding elements in the nuclear and the mitochondrial genome can generate dsRNAs. Here, the identity of endogenous dsRNAs, their recognition by dsRNA sensors, and their implications in the pathogenesis of human diseases ranging from inflammatory to degenerative are presented.


Assuntos
Doenças Autoimunes/imunologia , Imunidade Inata/imunologia , RNA de Cadeia Dupla/imunologia , RNA Mitocondrial/imunologia , Doenças Autoimunes/genética , Humanos , Conformação de Ácido Nucleico , RNA de Cadeia Dupla/genética , RNA Mitocondrial/genética , Transdução de Sinais/imunologia
12.
Biomacromolecules ; 21(6): 2440-2454, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32233463

RESUMO

Reactive poly(pentafluorophenyl acrylate) (PPFPA)-grafted surfaces offer a versatile platform to immobilize biomolecules. Here, we utilize PPFPA-grafted surface and double-stranded RNA (dsRNA) recognizing J2 antibody to construct a universal virus detection platform with enhanced sensitivity. PPFPA on silicon substrates is prepared, and surface hydrophilicity is modulated by partial substitution of the pentafluorophenyl units with poly(ethylene glycol). Following dsRNA antibody immobilization, the prepared surfaces can distinguish long dsRNAs from single-stranded RNAs of the same length and short dsRNAs. As long dsRNAs are common byproducts of viral transcription/replication, these surfaces can detect the presence of different kinds of viruses without prior knowledge of their genomic sequences. To increase dsRNA detection sensitivity, a two-step method is devised where the captured dsRNAs are visualized with multiple fluorophore-tagged J2 antibodies. We show that the developed platform can differentiate foreign long dsRNAs from cellular dsRNAs and other biomolecules present in the cell lysate. Moreover, when tested against cells infected with hepatitis A or C viruses, both viruses are successfully detected using a single platform. Our study shows that the developed PPFPA platform immobilized with J2 antibody can serve as a primary diagnostic tool to determine the infection status for a wide range of viruses.


Assuntos
Polímeros , RNA de Cadeia Dupla , RNA de Cadeia Dupla/genética
13.
Mol Cell ; 46(4): 384-6, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22633486

RESUMO

RNA silencing requires assembly of an effector complex, RNA-induced silencing complex (RISC), composed of a small RNA and the Ago protein. In this issue of Molecular Cell, Liu et al. (2012) provide insights of miRNP/RISC assembly pathways in mammals, using an in vitro human RISC assembly assay programmed by pre-miRNAs.

14.
Biomacromolecules ; 19(3): 951-961, 2018 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-29406744

RESUMO

Reactive pentafluorophenyl acrylate (PFPA) polymer brushes grafted on silica particles were prepared using surface-initiated reversible addition and fragmentation chain transfer polymerization. The polymer brush was successfully immobilized with antibody, then used for protein separation. The immunoprecipitated proteins showed successful enrichment of target protein, with reduced nonspecific background and less contamination from eluted antibodies. To further improve protein recovery, the hydrophobic poly(PFPA) brush was modified with hydrophilic poly(ethylene glycol) (PEG). The partially PEG-substituted poly(PFPA) brush showed better dispersion in aqueous solution, leading to improved antibody immobilization efficiency. By optimizing both the brush molecular weight and the degree of PEG substitution, an optimal balance between surface hydrophilicity and number of available PFP units was found, leading to efficient target protein purification. This study shows that poly(PFPA) platform offers a versatile approach to prepare biomolecule-activated surfaces with tunable surface property, which has potential applications in protein separation and other areas.


Assuntos
Anticorpos Antineoplásicos/química , Polímeros de Fluorcarboneto/química , Proteínas de Neoplasias/isolamento & purificação , Ácidos Polimetacrílicos/química , Células HeLa , Humanos , Proteínas de Neoplasias/química
15.
Development ; 139(21): 3962-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23048183

RESUMO

The transcriptional repressor Capicua (Cic) controls multiple aspects of Drosophila embryogenesis and has been implicated in vertebrate development and human diseases. Receptor tyrosine kinases (RTKs) can antagonize Cic-dependent gene repression, but the mechanisms responsible for this effect are not fully understood. Based on genetic and imaging studies in the early Drosophila embryo, we found that Torso RTK signaling can increase the rate of Cic degradation by changing its subcellular localization. We propose that Cic is degraded predominantly in the cytoplasm and show that Torso reduces the stability of Cic by controlling the rates of its nucleocytoplasmic transport. This model accounts for the experimentally observed spatiotemporal dynamics of Cic in the early embryo and might explain RTK-dependent control of Cic in other developmental contexts.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas HMGB/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Western Blotting , Padronização Corporal/genética , Padronização Corporal/fisiologia , Drosophila , Proteínas de Drosophila/genética , Feminino , Recuperação de Fluorescência Após Fotodegradação , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas HMGB/genética , Masculino , Receptores Proteína Tirosina Quinases/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
16.
Development ; 139(16): 3032-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22791891

RESUMO

The dorsoventral (DV) axis of the Drosophila embryo is patterned by a nuclear gradient of the Rel family transcription factor, Dorsal (Dl), that activates or represses numerous target genes in a region-specific manner. Here, we demonstrate that signaling by receptor tyrosine kinases (RTK) reduces nuclear levels and transcriptional activity of Dl, both at the poles and in the mid-body of the embryo. These effects depend on wntD, which encodes a Dl antagonist belonging to the Wingless/Wnt family of secreted factors. Specifically, we show that, via relief of Groucho- and Capicua-mediated repression, the Torso and EGFR RTK pathways induce expression of WntD, which in turn limits Dl nuclear localization at the poles and along the DV axis. Furthermore, this RTK-dependent control of Dl is important for restricting expression of its targets in both contexts. Thus, our results reveal a new mechanism of crosstalk, whereby RTK signals modulate the spatial distribution and activity of a developmental morphogen in vivo.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/genética , Padronização Corporal/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Mutação , Proteínas Nucleares/genética , Fosfoproteínas/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Peptídeos de Invertebrados/genética , Receptores de Peptídeos de Invertebrados/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética
17.
EMBO J ; 29(19): 3222-35, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20802461

RESUMO

ß-Arrestins have been implicated in the regulation of multiple signalling pathways. However, their role in organism development is not well understood. In this study, we report a new in vivo function of the Drosophila ß-arrestin Kurtz (Krz) in the regulation of two distinct developmental signalling modules: MAPK ERK and NF-κB, which transmit signals from the activated receptor tyrosine kinases (RTKs) and the Toll receptor, respectively. Analysis of the expression of effectors and target genes of Toll and the RTK Torso in krz maternal mutants reveals that Krz limits the activity of both pathways in the early embryo. Protein interaction studies suggest a previously uncharacterized mechanism for ERK inhibition: Krz can directly bind and sequester an inactive form of ERK, thus preventing its activation by the upstream kinase, MEK. A simultaneous dysregulation of different signalling systems in krz mutants results in an abnormal patterning of the embryo and severe developmental defects. Our findings uncover a new in vivo function of ß-arrestins and present a new mechanism of ERK inhibition by the Drosophila ß-arrestin Krz.


Assuntos
Arrestinas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Inibidores Enzimáticos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo , Animais , Arrestinas/farmacologia , Western Blotting , Células Cultivadas , Drosophila/metabolismo , Proteínas de Drosophila/farmacologia , Inibidores Enzimáticos/farmacologia , Técnicas de Inativação de Genes , Imunoprecipitação , Hibridização in Situ Fluorescente , Mutação/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Development ; 138(22): 4867-74, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22007136

RESUMO

A crucial issue in studies of morphogen gradients relates to their range: the distance over which they can act as direct regulators of cell signaling, gene expression and cell differentiation. To address this, we present a straightforward statistical framework that can be used in multiple developmental systems. We illustrate the developed approach by providing a point estimate and confidence interval for the spatial range of the graded distribution of nuclear Dorsal, a transcription factor that controls the dorsoventral pattern of the Drosophila embryo.


Assuntos
Bioestatística/métodos , Biologia Computacional , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Drosophila/metabolismo , Genes Controladores do Desenvolvimento , Morfogênese/genética , Animais , Fase de Clivagem do Zigoto/metabolismo , Biologia Computacional/métodos , Biologia Computacional/estatística & dados numéricos , Simulação por Computador , Drosophila/genética , Proteínas de Drosophila/análise , Proteínas de Drosophila/genética , Embrião não Mamífero/química , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Controladores do Desenvolvimento/fisiologia , Imageamento Tridimensional , Hibridização in Situ Fluorescente , Morfogênese/fisiologia , Concentração Osmolar , Distribuição Tecidual/genética
19.
Nat Methods ; 8(2): 171-176, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21186361

RESUMO

Quantitative studies of embryogenesis require the ability to monitor pattern formation and morphogenesis in large numbers of embryos, at multiple time points and in diverse genetic backgrounds. We describe a simple approach that greatly facilitates these tasks for Drosophila melanogaster embryos, one of the most advanced models of developmental genetics. Based on passive hydrodynamics, we developed a microfluidic embryo-trap array that can be used to rapidly order and vertically orient hundreds of embryos. We describe the physical principles of the design and used this platform to quantitatively analyze multiple morphogen gradients in the dorsoventral patterning system. Our approach can also be used for live imaging and, with slight modifications, could be adapted for studies of pattern formation and morphogenesis in other model organisms.


Assuntos
Drosophila melanogaster/química , Drosophila melanogaster/embriologia , Análise em Microsséries/métodos , Animais , Drosophila melanogaster/metabolismo , Análise em Microsséries/instrumentação , Transdução de Sinais
20.
Exp Mol Med ; 56(6): 1250-1262, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38871814

RESUMO

Alu elements are highly abundant primate-specific short interspersed nuclear elements that account for ~10% of the human genome. Due to their preferential location in gene-rich regions, especially in introns and 3' UTRs, Alu elements can exert regulatory effects on the expression of both host and neighboring genes. When two Alu elements with inverse orientations are positioned in close proximity, their transcription results in the generation of distinct double-stranded RNAs (dsRNAs), known as inverted Alu repeats (IRAlus). IRAlus are key immunogenic self-dsRNAs and post-transcriptional cis-regulatory elements that play a role in circular RNA biogenesis, as well as RNA transport and stability. Recently, IRAlus dsRNAs have emerged as regulators of transcription and activators of Z-DNA-binding proteins. The formation and activity of IRAlus can be modulated through RNA editing and interactions with RNA-binding proteins, and misregulation of IRAlus has been implicated in several immune-associated disorders. In this review, we summarize the emerging functions of IRAlus dsRNAs, the regulatory mechanisms governing IRAlus activity, and their relevance in the pathogenesis of human diseases.


Assuntos
Elementos Alu , Transcriptoma , Humanos , Elementos Alu/genética , Regulação da Expressão Gênica , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Animais , RNA Circular/genética , Sequências Repetidas Invertidas , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Edição de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA