Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.766
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 183(7): 2020-2035.e16, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33326746

RESUMEN

Thousands of proteins localize to the nucleus; however, it remains unclear which contain transcriptional effectors. Here, we develop HT-recruit, a pooled assay where protein libraries are recruited to a reporter, and their transcriptional effects are measured by sequencing. Using this approach, we measure gene silencing and activation for thousands of domains. We find a relationship between repressor function and evolutionary age for the KRAB domains, discover that Homeodomain repressor strength is collinear with Hox genetic organization, and identify activities for several domains of unknown function. Deep mutational scanning of the CRISPRi KRAB maps the co-repressor binding surface and identifies substitutions that improve stability/silencing. By tiling 238 proteins, we find repressors as short as ten amino acids. Finally, we report new activator domains, including a divergent KRAB. These results provide a resource of 600 human proteins containing effectors and demonstrate a scalable strategy for assigning functions to protein domains.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Sistemas CRISPR-Cas/genética , Femenino , Silenciador del Gen , Genes Reporteros , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células K562 , Lentivirus/fisiología , Anotación de Secuencia Molecular , Mutación/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Dominios Proteicos , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Transcripción Genética , Dedos de Zinc
2.
Cell ; 176(1-2): 198-212.e15, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30503211

RESUMEN

Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2ß forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2ß for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Células 3T3 , Animales , Carcinoma Basocelular/metabolismo , Línea Celular , Cromatina , Proteínas de Unión al ADN/fisiología , Células HEK293 , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiología , Histona Desacetilasa 1/metabolismo , Humanos , Proteínas de la Membrana/fisiología , Ratones , Chaperonas Moleculares/metabolismo , Lámina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1/fisiología , Dedos de Zinc
3.
Cell ; 173(2): 430-442.e17, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29606353

RESUMEN

Fetal hemoglobin (HbF, α2γ2) level is genetically controlled and modifies severity of adult hemoglobin (HbA, α2ß2) disorders, sickle cell disease, and ß-thalassemia. Common genetic variation affects expression of BCL11A, a regulator of HbF silencing. To uncover how BCL11A supports the developmental switch from γ- to ß- globin, we use a functional assay and protein binding microarray to establish a requirement for a zinc-finger cluster in BCL11A in repression and identify a preferred DNA recognition sequence. This motif appears in embryonic and fetal-expressed globin promoters and is duplicated in γ-globin promoters. The more distal of the duplicated motifs is mutated in individuals with hereditary persistence of HbF. Using the CUT&RUN approach to map protein binding sites in erythroid cells, we demonstrate BCL11A occupancy preferentially at the distal motif, which can be disrupted by editing the promoter. Our findings reveal that direct γ-globin gene promoter repression by BCL11A underlies hemoglobin switching.


Asunto(s)
Proteínas Portadoras/metabolismo , Hemoglobina Fetal/genética , Proteínas Nucleares/metabolismo , Secuencia de Bases , Sitios de Unión , Proteínas Portadoras/genética , Línea Celular , Cromatina/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Células Eritroides/citología , Células Eritroides/metabolismo , Edición Génica , Humanos , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Represoras , Dedos de Zinc/genética , Globinas beta/genética , Talasemia beta/genética , Talasemia beta/patología , gamma-Globinas/genética
4.
Nat Immunol ; 21(4): 422-433, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205880

RESUMEN

A20 is an anti-inflammatory protein that is strongly linked to human disease. Here, we find that mice expressing three distinct targeted mutations of A20's zinc finger 7 (ZF7) ubiquitin-binding motif uniformly developed digit arthritis with features common to psoriatic arthritis, while mice expressing point mutations in A20's OTU or ZF4 motifs did not exhibit this phenotype. Arthritis in A20ZF7 mice required T cells and MyD88, was exquisitely sensitive to tumor necrosis factor and interleukin-17A, and persisted in germ-free conditions. A20ZF7 cells exhibited prolonged IκB kinase activity that drove exaggerated transcription of late-phase nuclear factor-κB response genes in vitro and in prediseased mouse paws in vivo. In addition, mice expressing double-mutant A20 proteins in A20's ZF4 and ZF7 motifs died perinatally with multi-organ inflammation. Therefore, A20's ZF4 and ZF7 motifs synergistically prevent inflammatory disease in a non-catalytic manner.


Asunto(s)
Artritis Psoriásica/metabolismo , Inflamación/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Interleucina-17 , Ratones , Ratones Endogámicos C57BL , Mutación/genética , FN-kappa B/metabolismo , Unión Proteica/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación/fisiología , Dedos de Zinc/fisiología
5.
Nat Immunol ; 21(4): 381-387, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205881

RESUMEN

Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.


Asunto(s)
Inflamación/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Epiteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Poliubiquitina/metabolismo , Unión Proteica/fisiología , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Dedos de Zinc/fisiología
6.
Mol Cell ; 83(2): 252-265.e13, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36630955

RESUMEN

The conserved regulon of heat shock factor 1 in budding yeast contains chaperones for general protein folding as well as zinc-finger protein Zpr1, whose essential role in archaea and eukaryotes remains unknown. Here, we show that Zpr1 depletion causes acute proteotoxicity driven by biosynthesis of misfolded eukaryotic translation elongation factor 1A (eEF1A). Prolonged Zpr1 depletion leads to eEF1A insufficiency, thereby inducing the integrated stress response and inhibiting protein synthesis. Strikingly, we show by using two distinct biochemical reconstitution approaches that Zpr1 enables eEF1A to achieve a conformational state resistant to protease digestion. Lastly, we use a ColabFold model of the Zpr1-eEF1A complex to reveal a folding mechanism mediated by the Zpr1's zinc-finger and alpha-helical hairpin structures. Our work uncovers the long-sought-after function of Zpr1 as a bespoke chaperone tailored to the biogenesis of one of the most abundant proteins in the cell.


Asunto(s)
Proteínas Portadoras , Chaperonas Moleculares , Proteínas Portadoras/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Biosíntesis de Proteínas , Zinc/metabolismo , Dedos de Zinc , Factor 1 de Elongación Peptídica/metabolismo
7.
Genes Dev ; 37(15-16): 724-742, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37612136

RESUMEN

Histidine (His) residues are methylated in various proteins, but their roles and regulation mechanisms remain unknown. Here, we show that carnosine N-methyltransferase 1 (CARNMT1), a known His methyltransferase of dipeptide carnosine (ßAla-His), is a major His N1-position-specific methyltransferase. We found that 52 His sites in 20 proteins underwent CARNMT1-mediated methylation. The consensus methylation site for CARNMT1 was identified as Cx(F/Y)xH, a C3H zinc finger (C3H ZF) motif. CARNMT1-deficient and catalytically inactive mutant mice showed embryonic lethality. Among the CARNMT1 target C3H ZF proteins, RNA degradation mediated by Roquin and tristetraprolin (TTP) was affected by CARNMT1 and its enzymatic activity. Furthermore, the recognition of the 3' splice site of the CARNMT1 target C3H ZF protein U2AF1 was perturbed, and pre-mRNA alternative splicing (AS) was affected by CARNMT1 deficiency. These findings indicate that CARNMT1-mediated protein His methylation, which is essential for embryogenesis, plays roles in diverse aspects of RNA metabolism by targeting C3H ZF-type RNA-binding proteins and modulating their functions, including pre-mRNA AS and mRNA degradation regulation.


Asunto(s)
Carnosina , Animales , Ratones , Ratones Endogámicos C3H , Histidina/genética , Precursores del ARN , Metiltransferasas/genética , Sitios de Empalme de ARN , Dedos de Zinc
8.
Genes Dev ; 37(21-24): 984-997, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-37993255

RESUMEN

The RING-type E3 ligase has been known for over two decades, yet its diverse modes of action are still the subject of active research. Plant homeodomain (PHD) finger protein 7 (PHF7) is a RING-type E3 ubiquitin ligase responsible for histone ubiquitination. PHF7 comprises three zinc finger domains: an extended PHD (ePHD), a RING domain, and a PHD. While the function of the RING domain is largely understood, the roles of the other two domains in E3 ligase activity remain elusive. Here, we present the crystal structure of PHF7 in complex with the E2 ubiquitin-conjugating enzyme (E2). Our structure shows that E2 is effectively captured between the RING domain and the C-terminal PHD, facilitating E2 recruitment through direct contact. In addition, through in vitro binding and functional assays, we demonstrate that the N-terminal ePHD recognizes the nucleosome via DNA binding, whereas the C-terminal PHD is involved in histone H3 recognition. Our results provide a molecular basis for the E3 ligase activity of PHF7 and uncover the specific yet collaborative contributions of each domain to the PHF7 ubiquitination activity.


Asunto(s)
Histonas , Ubiquitina-Proteína Ligasas , Histonas/metabolismo , Ubiquitinación , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Dedos de Zinc , Enzimas Ubiquitina-Conjugadoras/metabolismo
9.
Immunity ; 54(6): 1168-1185.e8, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34038747

RESUMEN

Chronic inflammation plays a central role in hepatocellular carcinoma (HCC), but the contribution of hepatocytes to tumor-associated inflammation is not clear. Here, we report that the zinc finger transcription factor Miz1 restricted hepatocyte-driven inflammation to suppress HCC, independently of its transcriptional activity. Miz1 was downregulated in HCC mouse models and a substantial fraction of HCC patients. Hepatocyte-specific Miz1 deletion in mice generated a distinct sub-group of hepatocytes that produced pro-inflammatory cytokines and chemokines, which skewed the polarization of the tumor-infiltrating macrophages toward pro-inflammatory phenotypes to promote HCC. Mechanistically, Miz1 sequestrated the oncoprotein metadherin (MTDH), preventing MTDH from promoting transcription factor nuclear factor κB (NF-κB) activation. A distinct sub-group of pro-inflammatory cytokine-producing hepatocytes was also seen in a subset of HCC patients. In addition, Miz1 expression inversely correated with disease recurrence and poor prognosis in HCC patients. Our findings identify Miz1 as a tumor suppressor that prevents hepatocytes from driving inflammation in HCC.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma Hepatocelular/metabolismo , Hepatocitos/metabolismo , Inflamación/metabolismo , Neoplasias Hepáticas/metabolismo , Activación de Macrófagos/fisiología , Proteínas Inhibidoras de STAT Activados/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Quimiocinas/metabolismo , Regulación hacia Abajo/fisiología , Femenino , Células HEK293 , Hepatocitos/patología , Humanos , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Dedos de Zinc/fisiología
10.
Nature ; 627(8003): 416-423, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418872

RESUMEN

Permanent epigenetic silencing using programmable editors equipped with transcriptional repressors holds great promise for the treatment of human diseases1-3. However, to unlock its full therapeutic potential, an experimental confirmation of durable epigenetic silencing after the delivery of transient delivery of editors in vivo is needed. To this end, here we targeted Pcsk9, a gene expressed in hepatocytes that is involved in cholesterol homeostasis. In vitro screening of different editor designs indicated that zinc-finger proteins were the best-performing DNA-binding platform for efficient silencing of mouse Pcsk9. A single administration of lipid nanoparticles loaded with the editors' mRNAs almost halved the circulating levels of PCSK9 for nearly one year in mice. Notably, Pcsk9 silencing and accompanying epigenetic repressive marks also persisted after forced liver regeneration, further corroborating the heritability of the newly installed epigenetic state. Improvements in construct design resulted in the development of an all-in-one configuration that we term evolved engineered transcriptional repressor (EvoETR). This design, which is characterized by a high specificity profile, further reduced the circulating levels of PCSK9 in mice with an efficiency comparable with that obtained through conventional gene editing, but without causing DNA breaks. Our study lays the foundation for the development of in vivo therapeutics that are based on epigenetic silencing.


Asunto(s)
Epigénesis Genética , Epigenoma , Edición Génica , Silenciador del Gen , Animales , Ratones , Colesterol/metabolismo , Epigénesis Genética/genética , Epigenoma/genética , Edición Génica/métodos , Hepatocitos/metabolismo , Hígado/metabolismo , Regeneración Hepática , Nanopartículas , Proproteína Convertasa 9/sangre , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/genética , Proteínas Represoras/administración & dosificación , Proteínas Represoras/metabolismo , Dedos de Zinc
11.
Annu Rev Biochem ; 83: 409-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24606144

RESUMEN

Current technology enables the production of highly specific genome modifications with excellent efficiency and specificity. Key to this capability are targetable DNA cleavage reagents and cellular DNA repair pathways. The break made by these reagents can produce localized sequence changes through inaccurate nonhomologous end joining (NHEJ), often leading to gene inactivation. Alternatively, user-provided DNA can be used as a template for repair by homologous recombination (HR), leading to the introduction of desired sequence changes. This review describes three classes of targetable cleavage reagents: zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR/Cas RNA-guided nucleases (RGNs). As a group, these reagents have been successfully used to modify genomic sequences in a wide variety of cells and organisms, including humans. This review discusses the properties, advantages, and limitations of each system, as well as the specific considerations required for their use in different biological systems.


Asunto(s)
Endonucleasas/genética , Ingeniería Genética/métodos , Genoma , Animales , Arabidopsis , ADN/química , Daño del ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Drosophila , Drosophila melanogaster , Eliminación de Gen , Genómica , Humanos , Ratones , Ingeniería de Proteínas/métodos , Estructura Terciaria de Proteína , Ratas , Recombinación Genética , Pez Cebra , Dedos de Zinc
12.
Cell ; 157(5): 1061-72, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24855944

RESUMEN

TRPM7 is a ubiquitous ion channel and kinase, a unique "chanzyme," required for proper early embryonic development. It conducts Zn(2+), Mg(2+), and Ca(2+) as well as monovalent cations and contains a functional serine/threonine kinase at its carboxyl terminus. Here, we show that in normal tissues and cell lines, the kinase is proteolytically cleaved from the channel domain in a cell-type-specific manner. These TRPM7 cleaved kinase fragments (M7CKs) translocate to the nucleus and bind multiple components of chromatin-remodeling complexes, including Polycomb group proteins. In the nucleus, the kinase phosphorylates specific serines/threonines of histones. M7CK-dependent phosphorylation of H3Ser10 at promoters of TRPM7-dependent genes correlates with their activity. We also demonstrate that cytosolic free [Zn(2+)] is TRPM7 dependent and regulates M7CK binding to transcription factors containing zinc-finger domains. These findings suggest that TRPM7-mediated modulation of intracellular Zn(2+) concentration couples ion-channel signaling to epigenetic chromatin covalent modifications that affect gene expression patterns. PAPERCLIP:


Asunto(s)
Canales Catiónicos TRPM/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Citosol/metabolismo , Expresión Génica , Histonas/metabolismo , Humanos , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas , Zinc/metabolismo , Dedos de Zinc
13.
Mol Cell ; 81(2): 218-219, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33482089

RESUMEN

Lan et al. carry out a CRISPR-mediated genetic screen and discover that ZNF410 uniquely regulates the NuRD component CHD4 to repress γ-globin transcription in erythroid cells, establishing a novel fetal hemoglobin regulatory mechanism.


Asunto(s)
Hemoglobina Fetal , Factores de Transcripción , Células Eritroides , Hemoglobina Fetal/genética , Factores de Transcripción/genética , Dedos de Zinc/genética , gamma-Globinas
14.
Mol Cell ; 81(4): 845-858.e8, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33406384

RESUMEN

Mammalian genomes contain long domains with distinct average compositions of A/T versus G/C base pairs. In a screen for proteins that might interpret base composition by binding to AT-rich motifs, we identified the stem cell factor SALL4, which contains multiple zinc fingers. Mutation of the domain responsible for AT binding drastically reduced SALL4 genome occupancy and prematurely upregulated genes in proportion to their AT content. Inactivation of this single AT-binding zinc-finger cluster mimicked defects seen in Sall4 null cells, including precocious differentiation of embryonic stem cells (ESCs) and embryonic lethality in mice. In contrast, deletion of two other zinc-finger clusters was phenotypically neutral. Our data indicate that loss of pluripotency is triggered by downregulation of SALL4, leading to de-repression of a set of AT-rich genes that promotes neuronal differentiation. We conclude that base composition is not merely a passive byproduct of genome evolution and constitutes a signal that aids control of cell fate.


Asunto(s)
Composición de Base , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Ratones , Ratones Mutantes , Células Madre Embrionarias de Ratones/citología , Mutación , Neuronas/citología , Factores de Transcripción/genética , Regulación hacia Arriba , Dedos de Zinc
15.
Mol Cell ; 81(22): 4591-4604.e8, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34592134

RESUMEN

Protein ADP-ribosylation is a reversible post-translational modification that transfers ADP-ribose from NAD+ onto acceptor proteins. Poly(ADP-ribosyl)ation (PARylation), catalyzed by poly(ADP-ribose) polymerases (PARPs) and poly(ADP-ribose) glycohydrolases (PARGs), which remove the modification, regulates diverse cellular processes. However, the chemistry and physiological functions of mono(ADP-ribosyl)ation (MARylation) remain elusive. Here, we report that Arabidopsis zinc finger proteins SZF1 and SZF2, key regulators of immune gene expression, are MARylated by the noncanonical ADP-ribosyltransferase SRO2. Immune elicitation promotes MARylation of SZF1/SZF2 via dissociation from PARG1, which has an unconventional activity in hydrolyzing both poly(ADP-ribose) and mono(ADP-ribose) from acceptor proteins. MARylation antagonizes polyubiquitination of SZF1 mediated by the SH3 domain-containing proteins SH3P1/SH3P2, thereby stabilizing SZF1 proteins. Our study uncovers a noncanonical ADP-ribosyltransferase mediating MARylation of immune regulators and underpins the molecular mechanism of maintaining protein homeostasis by the counter-regulation of ADP-ribosylation and polyubiquitination to ensure proper immune responses.


Asunto(s)
ADP-Ribosilación , Proteínas de Arabidopsis/metabolismo , Arabidopsis/inmunología , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Inmunidad de la Planta , Ubiquitinación , Dedos de Zinc , ADP Ribosa Transferasas/metabolismo , Adenosina Difosfato/química , Arabidopsis/metabolismo , Sistemas CRISPR-Cas , Genes de Plantas , Glicósido Hidrolasas/metabolismo , Homeostasis , Humanos , Hidrólisis , Mutación , Plantas Modificadas Genéticamente , Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteostasis , Plantones/metabolismo , Especificidad por Sustrato , Tristetraprolina/química , Técnicas del Sistema de Dos Híbridos , Ubiquitina/química
16.
Nat Immunol ; 17(3): 259-68, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26808229

RESUMEN

The proinflammatory cytokines interleukin 12 (IL-12) and IL-23 connect innate responses and adaptive immune responses and are also involved in autoimmune and inflammatory diseases. Here we describe an epigenetic mechanism for regulation of the genes encoding IL-12 (Il12a and Il12b; collectively called 'Il12' here) and IL-23 (Il23a and Il12b; collectively called 'Il23' here) involving the deubiquitinase Trabid. Deletion of Zranb1 (which encodes Trabid) in dendritic cells inhibited induction of the expression of Il12 and Il23 by Toll-like receptors (TLRs), which impaired the differentiation of inflammatory T cells and protected mice from autoimmune inflammation. Trabid facilitated TLR-induced histone modifications at the promoters of Il12 and Il23, which involved deubiqutination and stabilization of the histone demethylase Jmjd2d. Our findings highlight an epigenetic mechanism for the regulation of Il12 and Il23 and establish Trabid as an innate immunological regulator of inflammatory T cell responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/genética , Epigénesis Genética , Interleucina-12/genética , Interleucina-23/genética , Proteasas Ubiquitina-Específicas/genética , Animales , Diferenciación Celular , Inmunoprecipitación de Cromatina , Encefalomielitis Autoinmune Experimental/inmunología , Citometría de Flujo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Immunoblotting , Inmunoprecipitación , Interleucina-12/inmunología , Interleucina-23/inmunología , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Receptores Toll-Like/metabolismo , Proteasas Ubiquitina-Específicas/inmunología , Dedos de Zinc/genética , Dedos de Zinc/inmunología
17.
Genes Dev ; 34(23-24): 1753-1761, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33122294

RESUMEN

Most eukaryotic pre-mRNAs must undergo 3'-end cleavage and polyadenylation prior to their export from the nucleus. A large number of proteins in several complexes participate in this 3'-end processing, including cleavage and polyadenylation specificity factor (CPSF) in mammals. The CPSF30 subunit contains five CCCH zinc fingers (ZFs), with ZF2-ZF3 being required for the recognition of the AAUAAA poly(A) signal. ZF4-ZF5 recruits the hFip1 subunit of CPSF, although the details of this interaction have not been characterized. Here we report the crystal structure of human CPSF30 ZF4-ZF5 in complex with residues 161-200 of hFip1 at 1.9 Å resolution, illuminating the molecular basis for their interaction. Unexpectedly, the structure reveals one hFip1 molecule binding to each ZF4 and ZF5, with a conserved mode of interaction. Our mutagenesis studies confirm that the CPSF30-hFip1 complex has 1:2 stoichiometry in vitro. Mutation of each binding site in CPSF30 still allows one copy of hFip1 to bind, while mutation of both sites abrogates binding. Our fluorescence polarization binding assays show that ZF4 has higher affinity for hFip1, with a Kd of 1.8 nM. We also demonstrate that two copies of the catalytic module of poly(A) polymerase (PAP) are recruited by the CPSF30-hFip1 complex in vitro, and both hFip1 binding sites in CPSF30 can support polyadenylation.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/química , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Modelos Moleculares , Factores de Escisión y Poliadenilación de ARNm/química , Sitios de Unión , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Escherichia coli/genética , Humanos , Mutación , Unión Proteica , Estructura Cuaternaria de Proteína , Dedos de Zinc/fisiología , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
18.
Annu Rev Genet ; 53: 393-416, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31518518

RESUMEN

Nearly half of the human genome consists of endogenous retroelements (EREs) and their genetic remnants, a small fraction of which carry the potential to propagate in the host genome, posing a threat to genome integrity and cell/organismal survival. The largest family of transcription factors in tetrapods, the Krüppel-associated box domain zinc finger proteins (KRAB-ZFPs), binds to specific EREs and represses their transcription. Since their first appearance over 400 million years ago, KRAB-ZFPs have undergone dramatic expansion and diversification in mammals, correlating with the invasions of new EREs. In this article we review our current understanding of the structure, function, and evolution of KRAB-ZFPs and discuss growing evidence that the arms race between KRAB-ZFPs and the EREs they target is a major driving force for the evolution of new traits in mammals, often accompanied by domestication of EREs themselves.


Asunto(s)
Inmunidad Celular/fisiología , Mamíferos/genética , Retroelementos , Dedos de Zinc/fisiología , Animales , Regulación de la Expresión Génica , Impresión Genómica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Meiosis , Familia de Multigenes , Dominios Proteicos
19.
Plant Cell ; 36(4): 919-940, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38180963

RESUMEN

Soil salinity results in oxidative stress and heavy losses to crop production. The S-acylated protein SALT TOLERANCE RECEPTOR-LIKE CYTOPLASMIC KINASE 1 (STRK1) phosphorylates and activates CATALASE C (CatC) to improve rice (Oryza sativa L.) salt tolerance, but the molecular mechanism underlying its S-acylation involved in salt signal transduction awaits elucidation. Here, we show that the DHHC-type zinc finger protein DHHC09 S-acylates STRK1 at Cys5, Cys10, and Cys14 and promotes salt and oxidative stress tolerance by enhancing rice H2O2-scavenging capacity. This modification determines STRK1 targeting to the plasma membrane or lipid nanodomains and is required for its function. DHHC09 promotes salt signaling from STRK1 to CatC via transphosphorylation, and its deficiency impairs salt signal transduction. Our findings demonstrate that DHHC09 S-acylates and anchors STRK1 to the plasma membrane to promote salt signaling from STRK1 to CatC, thereby regulating H2O2 homeostasis and improving salt stress tolerance in rice. Moreover, overexpression of DHHC09 in rice mitigates grain yield loss under salt stress. Together, these results shed light on the mechanism underlying the role of S-acylation in RLK/RLCK-mediated salt signal transduction and provide a strategy for breeding highly salt-tolerant rice.


Asunto(s)
Oryza , Tolerancia a la Sal , Tolerancia a la Sal/genética , Oryza/metabolismo , Peróxido de Hidrógeno/metabolismo , Homeostasis , Dedos de Zinc , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo
20.
Cell ; 150(3): 647-58, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863014

RESUMEN

Eukaryotic transcription factors (TFs) perform complex and combinatorial functions within transcriptional networks. Here, we present a synthetic framework for systematically constructing eukaryotic transcription functions using artificial zinc fingers, modular DNA-binding domains found within many eukaryotic TFs. Utilizing this platform, we construct a library of orthogonal synthetic transcription factors (sTFs) and use these to wire synthetic transcriptional circuits in yeast. We engineer complex functions, such as tunable output strength and transcriptional cooperativity, by rationally adjusting a decomposed set of key component properties, e.g., DNA specificity, affinity, promoter design, protein-protein interactions. We show that subtle perturbations to these properties can transform an individual sTF between distinct roles (activator, cooperative factor, inhibitory factor) within a transcriptional complex, thus drastically altering the signal processing behavior of multi-input systems. This platform provides new genetic components for synthetic biology and enables bottom-up approaches to understanding the design principles of eukaryotic transcriptional complexes and networks.


Asunto(s)
Redes Reguladoras de Genes , Saccharomyces cerevisiae/genética , Dedos de Zinc , Secuencia de Aminoácidos , Modelos Moleculares , Datos de Secuencia Molecular , Biología Sintética , Factores de Transcripción/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA