Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.356
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(2): 187-95, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26726812

RESUMEN

Studies of repertoires of mouse monoclonal CD4(+) T cells have revealed several mechanisms of self-tolerance; however, which mechanisms operate in normal repertoires is unclear. Here we studied polyclonal CD4(+) T cells specific for green fluorescent protein expressed in various organs, which allowed us to determine the effects of specific expression patterns on the same epitope-specific T cells. Peptides presented uniformly by thymic antigen-presenting cells were tolerated by clonal deletion, whereas peptides excluded from the thymus were ignored. Peptides with limited thymic expression induced partial clonal deletion and impaired effector T cell potential but enhanced regulatory T cell potential. These mechanisms were also active for T cell populations specific for endogenously expressed self antigens. Thus, the immunotolerance of polyclonal CD4(+) T cells was maintained by distinct mechanisms, according to self-peptide expression patterns.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Expresión Génica , Tolerancia Inmunológica , Péptidos/genética , Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Autoantígenos/química , Autoantígenos/genética , Autoantígenos/inmunología , Autoinmunidad , Supresión Clonal/genética , Supresión Clonal/inmunología , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Genes Reporteros , Ratones , Ratones Transgénicos , Péptidos/química , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/inmunología , Timo/metabolismo
2.
Nature ; 612(7941): 771-777, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36477533

RESUMEN

Human leucocyte antigen B*27 (HLA-B*27) is strongly associated with inflammatory diseases of the spine and pelvis (for example, ankylosing spondylitis (AS)) and the eye (that is, acute anterior uveitis (AAU))1. How HLA-B*27 facilitates disease remains unknown, but one possible mechanism could involve presentation of pathogenic peptides to CD8+ T cells. Here we isolated orphan T cell receptors (TCRs) expressing a disease-associated public ß-chain variable region-complementary-determining region 3ß (BV9-CDR3ß) motif2-4 from blood and synovial fluid T cells from individuals with AS and from the eye in individuals with AAU. These TCRs showed consistent α-chain variable region (AV21) chain pairing and were clonally expanded in the joint and eye. We used HLA-B*27:05 yeast display peptide libraries to identify shared self-peptides and microbial peptides that activated the AS- and AAU-derived TCRs. Structural analysis revealed that TCR cross-reactivity for peptide-MHC was rooted in a shared binding motif present in both self-antigens and microbial antigens that engages the BV9-CDR3ß TCRs. These findings support the hypothesis that microbial antigens and self-antigens could play a pathogenic role in HLA-B*27-associated disease.


Asunto(s)
Autoinmunidad , Antígenos HLA-B , Péptidos , Receptores de Antígenos de Linfocitos T , Humanos , Autoantígenos/química , Autoantígenos/inmunología , Autoantígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Antígenos HLA-B/inmunología , Antígenos HLA-B/metabolismo , Péptidos/química , Péptidos/inmunología , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Líquido Sinovial/inmunología , Espondilitis Anquilosante/inmunología , Uveítis Anterior/inmunología , Biblioteca de Péptidos , Reacciones Cruzadas , Secuencias de Aminoácidos
3.
Nat Immunol ; 16(11): 1153-61, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26437244

RESUMEN

Central to adaptive immunity is the interaction between the αß T cell receptor (TCR) and peptide presented by the major histocompatibility complex (MHC) molecule. Presumably reflecting TCR-MHC bias and T cell signaling constraints, the TCR universally adopts a canonical polarity atop the MHC. We report the structures of two TCRs, derived from human induced T regulatory (iT(reg)) cells, complexed to an MHC class II molecule presenting a proinsulin-derived peptide. The ternary complexes revealed a 180° polarity reversal compared to all other TCR-peptide-MHC complex structures. Namely, the iT(reg) TCR α-chain and ß-chain are overlaid with the α-chain and ß-chain of MHC class II, respectively. Nevertheless, this TCR interaction elicited a peptide-reactive, MHC-restricted T cell signal. Thus TCRs are not 'hardwired' to interact with MHC molecules in a stereotypic manner to elicit a T cell signal, a finding that fundamentally challenges our understanding of TCR recognition.


Asunto(s)
Autoantígenos/metabolismo , Complejo Mayor de Histocompatibilidad/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Adaptativa , Presentación de Antígeno , Autoantígenos/química , Autoantígenos/genética , Células Cultivadas , Antígeno HLA-DR4/química , Antígeno HLA-DR4/genética , Antígeno HLA-DR4/metabolismo , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Complejo Mayor de Histocompatibilidad/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proinsulina/química , Proinsulina/genética , Proinsulina/inmunología , Dominios y Motivos de Interacción de Proteínas , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T Reguladores/inmunología
4.
Cell ; 150(2): 317-26, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22817894

RESUMEN

In eukaryotes, DNA is packaged into chromatin by canonical histone proteins. The specialized histone H3 variant CENP-A provides an epigenetic and structural basis for chromosome segregation by replacing H3 at centromeres. Unlike exclusively octameric canonical H3 nucleosomes, CENP-A nucleosomes have been shown to exist as octamers, hexamers, and tetramers. An intriguing possibility reconciling these observations is that CENP-A nucleosomes cycle between octamers and tetramers in vivo. We tested this hypothesis by tracking CENP-A nucleosomal components, structure, chromatin folding, and covalent modifications across the human cell cycle. We report that CENP-A nucleosomes alter from tetramers to octamers before replication and revert to tetramers after replication. These structural transitions are accompanied by reversible chaperone binding, chromatin fiber folding changes, and previously undescribed modifications within the histone fold domains of CENP-A and H4. Our results reveal a cyclical nature to CENP-A nucleosome structure and have implications for the maintenance of epigenetic memory after centromere replication.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Nucleosomas/metabolismo , Autoantígenos/química , Ciclo Celular , Centrómero/metabolismo , Proteína A Centromérica , Proteínas Cromosómicas no Histona/química , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Células HeLa , Histonas/química , Histonas/metabolismo , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína
5.
Nat Immunol ; 15(2): 177-85, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24362891

RESUMEN

T cells autoreactive to the antigen-presenting molecule CD1a are common in human blood and skin, but the search for natural autoantigens has been confounded by background T cell responses to CD1 proteins and self lipids. After capturing CD1a-lipid complexes, we gently eluted ligands while preserving non-ligand-bound CD1a for testing lipids from tissues. CD1a released hundreds of ligands of two types. Inhibitory ligands were ubiquitous membrane lipids with polar head groups, whereas stimulatory compounds were apolar oils. We identified squalene and wax esters, which naturally accumulate in epidermis and sebum, as autoantigens presented by CD1a. The activation of T cells by skin oils suggested that headless mini-antigens nest within CD1a and displace non-antigenic resident lipids with large head groups. Oily autoantigens naturally coat the surface of the skin; thus, this points to a previously unknown mechanism of barrier immunity.


Asunto(s)
Antígenos CD1/inmunología , Autoantígenos/inmunología , Lípidos/inmunología , Piel/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Presentación de Antígeno , Antígenos CD1/genética , Autoantígenos/química , Autoantígenos/aislamiento & purificación , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lípidos/química , Lípidos/aislamiento & purificación , Activación de Linfocitos , Datos de Secuencia Molecular , Unión Proteica , Proteínas Recombinantes/genética , Relación Estructura-Actividad
6.
Nature ; 587(7835): 678-682, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32911480

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is an innate immune sensor for cytosolic microbial DNA1. After binding DNA, cGAS synthesizes the messenger 2'3'-cyclic GMP-AMP (cGAMP)2-4, which triggers cell-autonomous defence and the production of type I interferons and pro-inflammatory cytokines via the activation of STING5. In addition to responding to cytosolic microbial DNA, cGAS also recognizes mislocalized cytosolic self-DNA and has been implicated in autoimmunity and sterile inflammation6,7. Specificity towards pathogen- or damage-associated DNA was thought to be caused by cytosolic confinement. However, recent findings place cGAS robustly in the nucleus8-10, where tight tethering of chromatin is important to prevent autoreactivity to self-DNA8. Here we show how cGAS is sequestered and inhibited by chromatin. We provide a cryo-electron microscopy structure of the cGAS catalytic domain bound to a nucleosome, which shows that cGAS does not interact with the nucleosomal DNA, but instead interacts with histone 2A-histone 2B, and is tightly anchored to the 'acidic patch'. The interaction buries the cGAS DNA-binding site B, and blocks the formation of active cGAS dimers. The acidic patch robustly outcompetes agonistic DNA for binding to cGAS, which suggests that nucleosome sequestration can efficiently inhibit cGAS, even when accessible DNA is nearby, such as in actively transcribed genomic regions. Our results show how nuclear cGAS is sequestered by chromatin and provides a mechanism for preventing autoreactivity to nuclear self-DNA.


Asunto(s)
Dominio Catalítico , Cromatina/química , Cromatina/metabolismo , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/química , Secuencia de Aminoácidos , Animales , Autoantígenos/química , Autoantígenos/inmunología , Autoantígenos/metabolismo , Autoantígenos/ultraestructura , Sitios de Unión , Unión Competitiva , Cromatina/genética , Cromatina/ultraestructura , Microscopía por Crioelectrón , ADN/química , ADN/inmunología , ADN/metabolismo , ADN/ultraestructura , Activación Enzimática , Histonas/química , Histonas/metabolismo , Histonas/ultraestructura , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inmunidad Innata , Ratones , Modelos Moleculares , Nucleosomas/química , Nucleosomas/genética , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/ultraestructura , Multimerización de Proteína , Células THP-1
7.
Nature ; 568(7752): 344-350, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30944469

RESUMEN

Stem cells underlie tissue homeostasis, but their dynamics during ageing-and the relevance of these dynamics to organ ageing-remain unknown. Here we report that the expression of the hemidesmosome component collagen XVII (COL17A1) by epidermal stem cells fluctuates physiologically through genomic/oxidative stress-induced proteolysis, and that the resulting differential expression of COL17A1 in individual stem cells generates a driving force for cell competition. In vivo clonal analysis in mice and in vitro 3D modelling show that clones that express high levels of COL17A1, which divide symmetrically, outcompete and eliminate adjacent stressed clones that express low levels of COL17A1, which divide asymmetrically. Stem cells with higher potential or quality are thus selected for homeostasis, but their eventual loss of COL17A1 limits their competition, thereby causing ageing. The resultant hemidesmosome fragility and stem cell delamination deplete adjacent melanocytes and fibroblasts to promote skin ageing. Conversely, the forced maintenance of COL17A1 rescues skin organ ageing, thereby indicating potential angles for anti-ageing therapeutic intervention.


Asunto(s)
Homeostasis , Envejecimiento de la Piel/patología , Envejecimiento de la Piel/fisiología , Piel/citología , Piel/patología , Células Madre/citología , Células Madre/patología , Animales , Atrofia , Autoantígenos/química , Autoantígenos/metabolismo , División Celular , Proliferación Celular , Células Clonales/citología , Células Epidérmicas/citología , Células Epidérmicas/patología , Femenino , Genoma , Hemidesmosomas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Colágenos no Fibrilares/química , Colágenos no Fibrilares/metabolismo , Estrés Oxidativo , Proteolisis , Colágeno Tipo XVII
8.
Mol Cell ; 67(4): 646-658.e3, 2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28781232

RESUMEN

In miRNA-mediated gene silencing, the physical interaction between human Argonaute (hAgo) and GW182 (hGW182) is essential for facilitating the downstream silencing of the targeted mRNA. GW182 can interact with hAgo via three of the GW/WG repeats in its Argonaute-binding domain: motif-1, motif-2, and the hook motif. The structure of hAgo1 in complex with the hook motif of hGW182 reveals a "gate"-like interaction that is critical for GW182 docking into one of hAgo1's tryptophan-binding pockets. We show that hAgo1 and hAgo2 have a single GW182-binding site and that miRNA binding increases hAgo's affinity to GW182. With target binding occurring rapidly, this ensures that only mature RISC would be recruited for silencing. Finally, we show that hGW182 can recruit up to three copies of hAgo via its three GW motifs. This may explain the observed cooperativity in miRNA-mediated gene silencing.


Asunto(s)
Proteínas Argonautas/metabolismo , Autoantígenos/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Silenciador del Gen , MicroARNs/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Argonautas/química , Proteínas Argonautas/genética , Autoantígenos/química , Autoantígenos/genética , Sitios de Unión , Unión Competitiva , Cristalografía por Rayos X , Factores Eucarióticos de Iniciación/química , Factores Eucarióticos de Iniciación/genética , Humanos , MicroARNs/química , MicroARNs/genética , Simulación del Acoplamiento Molecular , Mutación , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Células Sf9 , Relación Estructura-Actividad , Transfección
9.
RNA Biol ; 21(1): 7-16, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39016322

RESUMEN

La-related proteins (LARPs) are a family of RNA-binding proteins that share a conserved La motif (LaM) domain. LARP1 plays a role in regulating ribosomal protein synthesis and stabilizing mRNAs and has a unique structure without an RNA binding RRM domain adjoining the LaM domain. In this study, we investigated the physical basis for LARP1 specificity for poly(A) sequences and observed an unexpected bias for sequences with single guanines. Multiple guanine substitutions did not increase the affinity, demonstrating preferential recognition of singly guanylated sequences. We also observed that the cyclic di-nucleotides in the cCAS/STING pathway, cyclic-di-GMP and 3',3'-cGAMP, bound with sub-micromolar affinity. Isothermal titration measurements were complemented by high-resolution crystal structures of the LARP1 LaM with six different RNA ligands, including two stereoisomers of a phosphorothioate linkage. The selectivity for singly substituted poly(A) sequences suggests LARP1 may play a role in the stabilizing effect of poly(A) tail guanylation. [Figure: see text].


Asunto(s)
Poli A , Unión Proteica , Ribonucleoproteínas , Antígeno SS-B , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/genética , Poli A/metabolismo , Poli A/química , Humanos , Modelos Moleculares , Sitios de Unión , Autoantígenos/metabolismo , Autoantígenos/química , Autoantígenos/genética , Cristalografía por Rayos X , Dominios Proteicos , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/química , ARN Mensajero/metabolismo , ARN Mensajero/química , ARN Mensajero/genética
10.
Genes Cells ; 27(9): 579-585, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35822830

RESUMEN

GW182 family proteins are a key component of microRNA-protein complex eliciting translational repression and/or degradation of microRNA-targets. The microRNAs in complex with Argonaute proteins bind to target mRNAs, and GW182 proteins are recruited by association with Argonaute proteins. The GW182 protein acts as a scaffold that links the Argonaute protein to silencing machineries including the CCR4-NOT complex which accelerates deadenylation and inhibits translation. The carboxyl-terminal effector domain of GW182 protein, also called the silencing domain, has been shown to bind to the subunits of the CCR4-NOT complex, the CNOT1 and the CNOT9. Here we show that a small region within the amino-terminal Argonaute-binding domain of human GW182/TNRC6A can associate with the CCR4-NOT complex. This region resides between the two Argonaute-binding sites and contains reiterated GW/WG-motifs. Alanine mutation experiments showed that multiple tryptophan residues are required for the association with the CCR4-NOT complex. Furthermore, co-expression and immunoprecipitation assays suggested that the CNOT9 subunit of the CCR4-NOT complex is a possible binding partner of this region. Our work, taken together with previous studies, indicates that the human GW182 protein contains multiple binding interfaces to the CCR4-NOT complex.


Asunto(s)
Proteínas Argonautas , Autoantígenos , MicroARNs , Proteínas de Unión al ARN , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Autoantígenos/química , Autoantígenos/genética , Autoantígenos/metabolismo , Sitios de Unión , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Unión Proteica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores CCR4/genética , Receptores CCR4/metabolismo , Factores de Transcripción/metabolismo , Triptófano/genética , Triptófano/metabolismo
11.
Blood ; 137(19): 2694-2698, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33544829

RESUMEN

Immune-mediated thrombotic thrombocytopenic purpura (iTTP) is an autoimmune disorder caused by the development of autoantibodies targeting different domains of ADAMTS13. Profiling studies have shown that residues R568, F592, R660, Y661, and Y665 within exosite-3 of the spacer domain provide an immunodominant region of ADAMTS13 for pathogenic autoantibodies that develop in patients with iTTP. Modification of these 5 core residues with the goal of reducing autoantibody binding revealed a significant tradeoff between autoantibody resistance and proteolytic activity. Here, we employed structural bioinformatics to identify a larger epitope landscape on the ADAMTS13 spacer domain. Models of spacer-antibody complexes predicted that residues R568, L591, F592, K608, M609, R636, L637, R639, R660, Y661, Y665, and L668 contribute to an expanded epitope within the spacer domain. Based on bioinformatics-guided predictions, we designed a panel of N-glycan insertions in this expanded epitope to reduce the binding of spacer domain autoantibodies. One N-glycan variant (NGLY3-ADAMTS13, containing a K608N substitution) showed strongly reduced reactivity with TTP patient sera (28%) as compared with WT-ADAMTS13 (100%). Insertion of an N-glycan at amino acid position 608 did not interfere with processing of von Willebrand factor, positioning the resulting NGLY3-ADAMTS13 variant as a potential novel therapeutic option for treatment of iTTP.


Asunto(s)
Proteína ADAMTS13/inmunología , Complejo Antígeno-Anticuerpo/química , Reacciones Antígeno-Anticuerpo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Polisacáridos/inmunología , Púrpura Trombocitopénica Trombótica/inmunología , Proteína ADAMTS13/química , Proteína ADAMTS13/metabolismo , Sustitución de Aminoácidos , Aminoácidos , Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Epítopos/inmunología , Epítopos/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios Proteicos , Factor de von Willebrand/metabolismo
12.
FASEB J ; 36(3): e22194, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35170814

RESUMEN

The leiomodin1 (LMOD1) gene, encoding a potent actin nucleator, was recently reported as a potential pathogenic gene of megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS, OMIM 619362). However, only a single patient has been reported to have LMOD1 mutations, and the underlying pathogenic mechanism remains unknown. Here, we described a male infant with LMOD1 mutations presenting typical symptoms of pediatric intestinal pseudo-obstruction (PIPO) but without megacystis and microcolon. Two compound heterozygous missense variants (c.1106C>T, p.T369M; c.1262G>A, p.R421H) were identified, both affecting highly conserved amino acid residues within the second actin-binding site (ABS2) domain of LMOD1. Expression analysis showed that both variants resulted in significantly reduced protein amounts, especially for p.T369M, which was almost undetectable. The reduction was only partially rescued by the proteasome inhibitor MG-132, indicating that there might be proteasome-independent pathways involved in the degradation of the mutant proteins. Molecular modeling showed that variant p.T369M impaired the local protein conformation of the ABS2 domain, while variant p.R421H directly impaired the intermolecular interaction between ABS2 and actin. Accordingly, both variants significantly damaged LMOD1-mediated actin nucleation. These findings provide further human genetic evidence supporting LMOD1 as a pathogenic gene underlying visceral myopathy including PIPO and MMIHS, strengthen the critical role of ABS2 domain in LMOD1-mediated actin nucleation, and moreover, reveal an unrecognized role of ABS2 in protein stability.


Asunto(s)
Actinas/metabolismo , Autoantígenos/genética , Proteínas del Citoesqueleto/genética , Seudoobstrucción Intestinal/genética , Mutación con Pérdida de Función , Autoantígenos/química , Autoantígenos/metabolismo , Sitios de Unión , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Células HeLa , Humanos , Lactante , Seudoobstrucción Intestinal/metabolismo , Seudoobstrucción Intestinal/patología , Masculino , Simulación del Acoplamiento Molecular , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estabilidad Proteica
13.
PLoS Biol ; 18(6): e3000679, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32555591

RESUMEN

Centriolar satellites are dynamic, membraneless granules composed of over 200 proteins. They store, modify, and traffic centrosome and primary cilium proteins, and help to regulate both the biogenesis and some functions of centrosomes and cilium. In most cell types, satellites cluster around the perinuclear centrosome, but their integrity and cellular distribution are dynamically remodeled in response to different stimuli, such as cell cycle cues. Dissecting the specific and temporal functions and mechanisms of satellites and how these are influenced by their cellular positioning and dynamics has been challenging using genetic approaches, particularly in ciliated and proliferating cells. To address this, we developed a chemical-based trafficking assay to rapidly and efficiently redistribute satellites to either the cell periphery or center, and fuse them into stable clusters in a temporally controlled way. Induced satellite clustering at either the periphery or center resulted in antagonistic changes in the pericentrosomal levels of a subset of proteins, revealing a direct and selective role for their positioning in protein targeting and sequestration. Systematic analysis of the interactome of peripheral satellite clusters revealed enrichment of proteins implicated in cilium biogenesis and mitosis. Importantly, induction of peripheral satellite targeting in ciliated cells revealed a function for satellites not just for efficient cilium assembly but also in the maintenance of steady-state cilia and in cilia disassembly by regulating the structural integrity of the ciliary axoneme. Finally, perturbing satellite distribution and dynamics inhibited their mitotic dissolution, and mitotic progression was perturbed only in cells with centrosomal satellite clustering. Collectively, our results for the first time showed a direct link between satellite functions and their pericentrosomal clustering, suggested new mechanisms underlying satellite functions during cilium assembly, and provided a new tool for probing temporal satellite functions in different contexts.


Asunto(s)
Centriolos/metabolismo , Cilios/metabolismo , Gránulos Citoplasmáticos/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Mitosis , Fenotipo , Dominios Proteicos , Multimerización de Proteína , Reproducibilidad de los Resultados
14.
Nucleic Acids Res ; 49(20): 11596-11613, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34723322

RESUMEN

Using the programmable RNA-sequence binding domain of the Pumilio protein, we FLAG-tagged Xist (inactivated X chromosome specific transcript) in live mouse cells. Affinity pulldown coupled to mass spectrometry was employed to identify a list of 138 candidate Xist-binding proteins, from which, Ssb (also known as the lupus autoantigen La) was validated as a protein functionally critical for X chromosome inactivation (XCI). Extensive XCI defects were detected in Ssb knockdown cells, including chromatin compaction, death of female mouse embryonic stem cells during in vitro differentiation and chromosome-wide monoallelic gene expression pattern. Live-cell imaging of Xist RNA reveals the defining XCI defect: Xist cloud formation. Ssb is a ubiquitous and versatile RNA-binding protein with RNA chaperone and RNA helicase activities. Functional dissection of Ssb shows that the RNA chaperone domain plays critical roles in XCI. In Ssb knockdown cells, Xist transcripts are unstable and misfolded. These results show that Ssb is critically involved in XCI, possibly as a protein regulating the in-cell structure of Xist.


Asunto(s)
Pliegue del ARN , ARN Largo no Codificante/química , Proteínas de Unión al ARN/metabolismo , Inactivación del Cromosoma X , Animales , Autoantígenos/química , Autoantígenos/metabolismo , Sitios de Unión , Línea Celular , Ratones , Unión Proteica , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética
15.
Nucleic Acids Res ; 49(6): 3461-3489, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33398329

RESUMEN

LARP1 is a key repressor of TOP mRNA translation. It binds the m7Gppp cap moiety and the adjacent 5'TOP motif of TOP mRNAs, thus impeding the assembly of the eIF4F complex on these transcripts. mTORC1 controls TOP mRNA translation via LARP1, but the details of the mechanism are unclear. Herein we elucidate the mechanism by which mTORC1 controls LARP1's translation repression activity. We demonstrate that mTORC1 phosphorylates LARP1 in vitro and in vivo, activities that are efficiently inhibited by rapamycin and torin1. We uncover 26 rapamycin-sensitive phospho-serine and -threonine residues on LARP1 that are distributed in 7 clusters. Our data show that phosphorylation of a cluster of residues located proximally to the m7Gppp cap-binding DM15 region is particularly sensitive to rapamycin and regulates both the RNA-binding and the translation inhibitory activities of LARP1. Our results unravel a new model of translation control in which the La module (LaMod) and DM15 region of LARP1, both of which can directly interact with TOP mRNA, are differentially regulated: the LaMod remains constitutively bound to PABP (irrespective of the activation status of mTORC1), while the C-terminal DM15 'pendular hook' engages the TOP mRNA 5'-end to repress translation, but only in conditions of mTORC1 inhibition.


Asunto(s)
Autoantígenos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Biosíntesis de Proteínas , Ribonucleoproteínas/metabolismo , Secuencias de Aminoácidos , Autoantígenos/química , Células HEK293 , Humanos , Naftiridinas/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica , Ribonucleoproteínas/química , Serina/metabolismo , Sirolimus/farmacología , Treonina/metabolismo , Tirosina/metabolismo , Antígeno SS-B
16.
Proc Natl Acad Sci U S A ; 117(36): 22341-22350, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32855302

RESUMEN

Conformational diversity and self-cross-reactivity of antigens have been correlated with evasion from neutralizing antibody responses. We utilized single cell B cell sequencing, biolayer interferometry and X-ray crystallography to trace mutation selection pathways where the antibody response must resolve cross-reactivity between foreign and self-proteins bearing near-identical contact surfaces, but differing in conformational flexibility. Recurring antibody mutation trajectories mediate long-range rearrangements of framework (FW) and complementarity determining regions (CDRs) that increase binding site conformational diversity. These antibody mutations decrease affinity for self-antigen 19-fold and increase foreign affinity 67-fold, to yield a more than 1,250-fold increase in binding discrimination. These results demonstrate how conformational diversity in antigen and antibody does not act as a barrier, as previously suggested, but rather facilitates high affinity and high discrimination between foreign and self.


Asunto(s)
Anticuerpos , Diversidad de Anticuerpos/genética , Autoantígenos , Reordenamiento Génico de Linfocito B/genética , Mutación/genética , Animales , Anticuerpos/química , Anticuerpos/genética , Anticuerpos/metabolismo , Afinidad de Anticuerpos/genética , Autoanticuerpos/química , Autoanticuerpos/genética , Autoanticuerpos/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Regiones Determinantes de Complementariedad/genética , Inmunidad Humoral/genética , Ratones , Modelos Moleculares , Conformación Proteica , Hipermutación Somática de Inmunoglobulina/genética
17.
Genes Dev ; 29(22): 2337-42, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26543159

RESUMEN

NRMT1 is an N-terminal methyltransferase that methylates histone CENP-A as well as nonhistone substrates. Here, we report the crystal structure of human NRMT1 bound to CENP-A peptide at 1.3 Å. NRMT1 adopts a core methyltransferase fold that resembles DOT1L and PRMT but not SET domain family histone methyltransferases. Key substrate recognition and catalytic residues were identified by mutagenesis studies. Histone peptide profiling revealed that human NRMT1 is highly selective to human CENP-A and fruit fly H2B, which share a common "Xaa-Pro-Lys/Arg" motif. These results, along with a 1.5 Å costructure of human NRMT1 bound to the fruit fly H2B peptide, underscore the importance of the NRMT1 recognition motif.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Metiltransferasas/metabolismo , Modelos Moleculares , Animales , Autoantígenos/química , Proteína A Centromérica , Proteínas Cromosómicas no Histona/química , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Escherichia coli/genética , Histonas/química , Histonas/metabolismo , Metilación , Metiltransferasas/genética , Mutagénesis , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Int J Mol Sci ; 24(21)2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37958853

RESUMEN

Centromeric chromatin is thought to play a critical role in ensuring the faithful segregation of chromosomes during mitosis. However, our understanding of this role is presently limited by our poor understanding of the structure and composition of this unique chromatin. The nucleosomal variant, CENP-A, localizes to narrow regions within the centromere, where it plays a major role in centromeric function, effectively serving as a platform on which the kinetochore is assembled. Previous work found that, within a given cell, the number of microtubules within kinetochores is essentially unchanged between CENP-A-localized regions of different physical sizes. However, it is unknown if the amount of CENP-A is also unchanged between these regions of different sizes, which would reflect a strict structural correspondence between these two key characteristics of the centromere/kinetochore assembly. Here, we used super-resolution optical microscopy to image and quantify the amount of CENP-A and DNA within human centromere chromatin. We found that the amount of CENP-A within CENP-A domains of different physical sizes is indeed the same. Further, our measurements suggest that the ratio of CENP-A- to H3-containing nucleosomes within these domains is between 8:1 and 11:1. Thus, our results not only identify an unexpectedly strict relationship between CENP-A and microtubules stoichiometries but also that the CENP-A centromeric domain is almost exclusively composed of CENP-A nucleosomes.


Asunto(s)
Microscopía , Nucleosomas , Humanos , Proteína A Centromérica/genética , Proteínas Cromosómicas no Histona/metabolismo , Centrómero/metabolismo , Cromatina , Cinetocoros/metabolismo , Autoantígenos/química
19.
Nat Rev Mol Cell Biol ; 11(5): 379-84, 2010 05.
Artículo en Inglés | MEDLINE | ID: mdl-20379206

RESUMEN

GW182 proteins have emerged as key components of microRNA (miRNA) silencing complexes in animals. Although the precise molecular function of GW182 proteins is not fully understood, new findings indicate that they act as poly(A)-binding protein (PABP)-interacting proteins (PAIPs) that promote gene silencing, at least in part, by interfering with cytoplasmic PABP1 (PABPC1) function during translation and mRNA stabilization. This recent discovery paves the way for future studies of miRNA silencing mechanisms.


Asunto(s)
Autoantígenos/metabolismo , MicroARNs/metabolismo , Proteína I de Unión a Poli(A)/metabolismo , Secuencia de Aminoácidos , Animales , Autoantígenos/química , Sitios de Unión , Silenciador del Gen , Datos de Secuencia Molecular , Proteína I de Unión a Poli(A)/antagonistas & inhibidores , Unión Proteica , Estructura Terciaria de Proteína
20.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36232691

RESUMEN

CENP-A is a histone variant found in high abundance at the centromere in humans. At the centromere, this histone variant replaces the histone H3 found throughout the bulk chromatin. Additionally, the centromere comprises tandem repeats of α-satellite DNA, which CENP-A nucleosomes assemble upon. However, the effect of the DNA sequence on the nucleosome assembly and centromere formation remains poorly understood. Here, we investigated the structure of nucleosomes assembled with the CENP-A variant using Atomic Force Microscopy. We assembled both CENP-A nucleosomes and H3 nucleosomes on a DNA substrate containing an α-satellite motif and characterized their positioning and wrapping efficiency. We also studied CENP-A nucleosomes on the 601-positioning motif and non-specific DNA to compare their relative positioning and stability. CENP-A nucleosomes assembled on α-satellite DNA did not show any positional preference along the substrate, which is similar to both H3 nucleosomes and CENP-A nucleosomes on non-specific DNA. The range of nucleosome wrapping efficiency was narrower on α-satellite DNA compared with non-specific DNA, suggesting a more stable complex. These findings indicate that DNA sequence and histone composition may be two of many factors required for accurate centromere assembly.


Asunto(s)
División del Núcleo Celular , Proteína A Centromérica , Centrómero , ADN , Histonas , Nucleosomas , Autoantígenos/química , Autoantígenos/genética , División del Núcleo Celular/genética , División del Núcleo Celular/fisiología , Centrómero/genética , Centrómero/metabolismo , Proteína A Centromérica/genética , Proteína A Centromérica/metabolismo , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN/química , ADN/genética , ADN/metabolismo , ADN Satélite , Histonas/genética , Histonas/metabolismo , Humanos , Microscopía de Fuerza Atómica , Nucleosomas/genética , Nucleosomas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA