Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Struct Mol Biol ; 31(4): 633-643, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38267599

RESUMEN

Pioneer transcription factors are vital for cell fate changes. PU.1 and C/EBPα work together to regulate hematopoietic stem cell differentiation. However, how they recognize in vivo nucleosomal DNA targets remains elusive. Here we report the structures of the nucleosome containing the mouse genomic CX3CR1 enhancer DNA and its complexes with PU.1 alone and with both PU.1 and the C/EBPα DNA binding domain. Our structures reveal that PU.1 binds the DNA motif at the exit linker, shifting 17 bp of DNA into the core region through interactions with H2A, unwrapping ~20 bp of nucleosomal DNA. C/EBPα binding, aided by PU.1's repositioning, unwraps ~25 bp of entry DNA. The PU.1 Q218H mutation, linked to acute myeloid leukemia, disrupts PU.1-H2A interactions. PU.1 and C/EBPα jointly displace linker histone H1 and open the H1-condensed nucleosome array. Our study unveils how two pioneer factors can work cooperatively to open closed chromatin by altering DNA positioning in the nucleosome.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT , Nucleosomas , Ratones , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ADN/química
2.
bioRxiv ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37986743

RESUMEN

Pioneer transcription factors possess the unique ability to access DNA within tightly packed chromatin structures, playing pivotal roles in cell differentiation and reprogramming. However, their precise mechanism for recognizing nucleosomes has remained mystery. Recent structural and biochemical investigations into the binding interactions between the human pioneer factor OCT4 and the LIN28B nucleosome by Sinha et al.1 and Guan et al.2 have yielded conflicting results regarding nucleosome positioning, nucleosomal DNA unwrapping, binding cooperativity, and the role of N-terminal tail of OCT4. In this study, we undertook a comparative analysis of these two research efforts and delved into the factors contributing to the observed discrepancies. Our investigation unveiled that the utilization of human and Xenopus laevis core histones, along with a discrete two-step salt dialysis method, led to distinct positioning of DNA within reconstituted LIN28B nucleosomes. Additionally, our reanalysis of the electrophoretic mobility shift assay data showed that H3 K27 acetylation did not increase OCT4 binding to the internal sites of the nucleosome when normalized to input; instead, it promoted sample aggregation. Thus, the available experimental data support the notion that the human LIN28B nucleosome is pre-positioned for efficient binding with multiple OCT4s, and there is no compelling evidence for its regulation by histone modifications.

3.
bioRxiv ; 2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37790476

RESUMEN

Pioneer transcription factors are vital for cell fate changes. PU.1 and C/EBPα work together to regulate hematopoietic stem cell differentiation. However, how they recognize in vivo nucleosomal DNA targets remain elusive. Here we report the structures of the nucleosome containing the mouse genomic CX3CR1 enhancer DNA and its complexes with PU.1 alone and with both PU.1 and the C/EBPα DNA binding domain. Our structures reveal that PU.1 binds the DNA motif at the exit linker, shifting 17 bp of DNA into the core region through interactions with H2A, unwrapping ~20 bp of nucleosomal DNA. C/EBPα binding, aided by PU.1's repositioning, unwraps ~25 bp entry DNA. The PU.1 Q218H mutation, linked to acute myeloid leukemia, disrupts PU.1-H2A interactions. PU.1 and C/EBPα jointly displace linker histone H1 and open the H1-condensed nucleosome array. Our study unveils how two pioneer factors can work cooperatively to open closed chromatin by altering DNA positioning in the nucleosome.

4.
Nat Struct Mol Biol ; 30(11): 1794-1805, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37845412

RESUMEN

Organic anion transporters (OATs) of the SLC22 family have crucial roles in the transport of organic anions, including metabolites and therapeutic drugs, and in transporter-mediated drug-drug interactions. In the kidneys, OATs facilitate the elimination of metabolic waste products and xenobiotics. However, their transport activities can lead to the accumulation of certain toxic compounds within cells, causing kidney damage. Moreover, OATs are important drug targets, because their inhibition modulates the elimination or retention of substrates linked to diseases. Despite extensive research on OATs, the molecular basis of their substrate and inhibitor binding remains poorly understood. Here we report the cryo-EM structures of rat OAT1 (also known as SLC22A6) and its complexes with para-aminohippuric acid and probenecid at 2.1, 2.8 and 2.9 Å resolution, respectively. Our findings reveal a highly conserved substrate binding mechanism for SLC22 transporters, wherein four aromatic residues form a cage to accommodate the polyspecific binding of diverse compounds.


Asunto(s)
Proteína 1 de Transporte de Anión Orgánico , Transportadores de Anión Orgánico , Ratas , Animales , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Microscopía por Crioelectrón , Proteínas de Transporte de Membrana/metabolismo , Transportadores de Anión Orgánico/metabolismo , Riñón/metabolismo
5.
Mol Cell ; 83(12): 1970-1982.e6, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37327775

RESUMEN

Pioneer transcription factors are essential for cell fate changes by targeting closed chromatin. OCT4 is a crucial pioneer factor that can induce cell reprogramming. However, the structural basis of how pioneer factors recognize the in vivo nucleosomal DNA targets is unknown. Here, we determine the high-resolution structures of the nucleosome containing human LIN28B DNA and its complexes with the OCT4 DNA binding region. Three OCT4s bind the pre-positioned nucleosome by recognizing non-canonical DNA sequences. Two use their POUS domains while the other uses the POUS-loop-POUHD region; POUHD serves as a wedge to unwrap ∼25 base pair DNA. Our analysis of previous genomic data and determination of the ESRRB-nucleosome-OCT4 structure confirmed the generality of these structural features. Moreover, biochemical studies suggest that multiple OCT4s cooperatively open the H1-condensed nucleosome array containing the LIN28B nucleosome. Thus, our study suggests a mechanism of how OCT4 can target the nucleosome and open closed chromatin.


Asunto(s)
Cromatina , Nucleosomas , Factor 3 de Transcripción de Unión a Octámeros , Proteínas de Unión al ARN , Humanos , Secuencia de Bases , Reprogramación Celular , Cromatina/genética , ADN/metabolismo , Nucleosomas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
6.
bioRxiv ; 2023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-36789416

RESUMEN

Pioneer transcription factors are essential for cell fate changes by targeting closed chromatin. OCT4 is a crucial pioneer factor that can induce cell reprogramming. However, the structural basis of how pioneer factors recognize the in vivo nucleosomal DNA targets is unknown. Here, we determine the high-resolution structures of the nucleosome containing human LIN28B DNA and its complexes with the OCT4 DNA binding region. Three OCT4s bind the pre-positioned nucleosome by recognizing non-canonical DNA motifs. Two use their POUS domains by forming extensive hydrogen bonds. The other uses the POUS-loop-POUHD region; POUHD serves as a wedge to unwrap ∻25 base pair DNA. Biochemical studies suggest that multiple OCT4s cooperatively open the H1-condensed nucleosome array containing the LIN28B nucleosome. Our study suggests a mechanism whereby OCT4s target the LIN28B nucleosome by forming multivalent interactions with nucleosomal motifs, unwrapping nucleosomal DNA, evicting H1, and cooperatively open closed chromatin to initiate cell reprogramming.

7.
Dev Cell ; 57(5): 670-685.e8, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35231446

RESUMEN

The dually lipidated Sonic hedgehog (SHH) morphogen signals through the tumor suppressor membrane protein Patched1 (PTCH1) to activate the Hedgehog pathway, which is fundamental in development and cancer. SHH engagement with PTCH1 requires the GAS1 coreceptor, but the mechanism is unknown. We demonstrate a unique role for GAS1, catalyzing SHH-PTCH1 complex assembly in vertebrate cells by direct SHH transfer from the extracellular SCUBE2 carrier to PTCH1. Structure of the GAS1-SHH-PTCH1 transition state identifies how GAS1 recognizes the SHH palmitate and cholesterol modifications in modular fashion and how it facilitates lipid-dependent SHH handoff to PTCH1. Structure-guided experiments elucidate SHH movement from SCUBE2 to PTCH1, explain disease mutations, and demonstrate that SHH-induced PTCH1 dimerization causes its internalization from the cell surface. These results define how the signaling-competent SHH-PTCH1 complex assembles, the key step triggering the Hedgehog pathway, and provide a paradigm for understanding morphogen reception and its regulation.


Asunto(s)
Proteínas Hedgehog , Receptor Patched-1 , Transducción de Señal , Catálisis , Colesterol/metabolismo , Proteínas Hedgehog/metabolismo , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Relación Estructura-Actividad
8.
Nat Commun ; 12(1): 1763, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741944

RESUMEN

Accurate chromosome segregation relies on the specific centromeric nucleosome-kinetochore interface. In budding yeast, the centromere CBF3 complex guides the deposition of CENP-A, an H3 variant, to form the centromeric nucleosome in a DNA sequence-dependent manner. Here, we determine the structures of the centromeric nucleosome containing the native CEN3 DNA and the CBF3core bound to the canonical nucleosome containing an engineered CEN3 DNA. The centromeric nucleosome core structure contains 115 base pair DNA including a CCG motif. The CBF3core specifically recognizes the nucleosomal CCG motif through the Gal4 domain while allosterically altering the DNA conformation. Cryo-EM, modeling, and mutational studies reveal that the CBF3core forms dynamic interactions with core histones H2B and CENP-A in the CEN3 nucleosome. Our results provide insights into the structure of the budding yeast centromeric nucleosome and the mechanism of its assembly, which have implications for analogous processes of human centromeric nucleosome formation.


Asunto(s)
Centrómero/metabolismo , ADN de Hongos/metabolismo , Proteínas de Unión al ADN/metabolismo , Cinetocoros/metabolismo , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Centrómero/genética , Centrómero/ultraestructura , Proteína A Centromérica/química , Proteína A Centromérica/genética , Proteína A Centromérica/metabolismo , Microscopía por Crioelectrón , ADN de Hongos/química , ADN de Hongos/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Histonas/química , Histonas/genética , Histonas/metabolismo , Humanos , Cinetocoros/química , Conformación de Ácido Nucleico , Nucleosomas/genética , Nucleosomas/ultraestructura , Unión Proteica , Dominios Proteicos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Homología de Secuencia de Aminoácido
9.
Nat Microbiol ; 6(3): 380-391, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33452495

RESUMEN

Plasmodium falciparum VAR2CSA binds to chondroitin sulfate A (CSA) on the surface of the syncytiotrophoblast during placental malaria. This interaction facilitates placental sequestration of malaria parasites resulting in severe health outcomes for both the mother and her offspring. Furthermore, CSA is presented by diverse cancer cells and specific targeting of cells by VAR2CSA may become a viable approach for cancer treatment. In the present study, we determined the cryo-electron microscopy structures of the full-length ectodomain of VAR2CSA from P. falciparum strain NF54 in complex with CSA, and VAR2CSA from a second P. falciparum strain FCR3. The architecture of VAR2CSA is composed of a stable core flanked by a flexible arm. CSA traverses the core domain by binding within two channels and CSA binding does not induce major conformational changes in VAR2CSA. The CSA-binding elements are conserved across VAR2CSA variants and are flanked by polymorphic segments, suggesting immune selection outside the CSA-binding sites. This work provides paths for developing interventions against placental malaria and cancer.


Asunto(s)
Antígenos de Protozoos/metabolismo , Sulfatos de Condroitina/metabolismo , Placenta/metabolismo , Plasmodium falciparum/metabolismo , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Sitios de Unión , Sulfatos de Condroitina/química , Microscopía por Crioelectrón , Epítopos , Femenino , Variación Genética , Humanos , Vacunas contra la Malaria , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Placenta/parasitología , Plasmodium falciparum/química , Plasmodium falciparum/inmunología , Embarazo , Complicaciones Parasitarias del Embarazo , Unión Proteica , Dominios Proteicos
10.
Autophagy ; 16(12): 2292-2293, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33016201

RESUMEN

ATG9, the only transmembrane protein in the core macroautophagy/autophagy machinery, is a key player in the early stages of autophagosome formation. Yet, the lack of a high-resolution structure of ATG9 was a major impediment in understanding its three-dimensional organization and function. We recently solved a high-resolution cryoEM structure of the ubiquitously expressed human ATG9A isoform. The structure revealed that ATG9A is a domain-swapped homotrimer with a unique fold, and has an internal network of branched cavities. In cellulo analyses demonstrated the functional importance of the cavity-lining residues. These cavities could serve as conduits for transport of hydrophilic moieties, such as lipid headgroups, across the bilayer. Finally, structure-guided molecular dynamics predicted that ATG9A has membrane-bending properties, which is consistent with its localization to highly curved membranes.


Asunto(s)
Autofagia , Membrana Dobles de Lípidos , Proteínas Relacionadas con la Autofagia , Humanos , Proteínas de la Membrana , Proteínas de Transporte Vesicular
11.
Cell Rep ; 31(13): 107837, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32610138

RESUMEN

Autophagy is a catabolic process involving capture of cytoplasmic materials into double-membraned autophagosomes that subsequently fuse with lysosomes for degradation of the materials by lysosomal hydrolases. One of the least understood components of the autophagy machinery is the transmembrane protein ATG9. Here, we report a cryoelectron microscopy structure of the human ATG9A isoform at 2.9-Å resolution. The structure reveals a fold with a homotrimeric domain-swapped architecture, multiple membrane spans, and a network of branched cavities, consistent with ATG9A being a membrane transporter. Mutational analyses support a role for the cavities in the function of ATG9A. In addition, structure-guided molecular simulations predict that ATG9A causes membrane bending, explaining the localization of this protein to small vesicles and highly curved edges of growing autophagosomes.


Asunto(s)
Proteínas Relacionadas con la Autofagia/química , Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Proteínas Relacionadas con la Autofagia/ultraestructura , Microscopía por Crioelectrón , Células HEK293 , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/química , Proteínas de la Membrana/ultraestructura , Simulación de Dinámica Molecular , Mutagénesis/genética , Fosfatidilcolinas/química , Dominios Proteicos , Multimerización de Proteína , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas de Transporte Vesicular/ultraestructura
12.
Cell Rep ; 19(7): 1334-1342, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28514654

RESUMEN

Jasmonates (JAs) are essential plant hormones that play important roles in the regulation of plant growth and the response to environmental stress. In the JA signaling pathway, the core transcription factors are a class of basic helix-loop-helix (bHLH) proteins, including MYC2, MYC3, and MYC4, that have different regulatory capacities. Here, we report the 2.7 Å crystal structure of the MYC2 bHLH domain complexed with G-box DNA, showing a cis-tetrameric structure. Biochemical assays confirmed that full-length MYC2 forms a stable homo-tetramer both in solution and in DNA-bound states, whereas MYC3 forms only a homodimer. Isothermal titration calorimetry (ITC) assays demonstrated that tetramerization enhanced DNA binding affinity, and fluorescence resonance energy transfer (FRET) assay indicated DNA looping potential of tetrameric MYC2. Luciferase assay further confirmed the importance of tetramerization in transcriptional regulation. Our studies provide a mechanistic explanation for the regulatory differences of MYC transcription factors.


Asunto(s)
Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/química , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , ADN de Plantas/metabolismo , Multimerización de Proteína , Secuencia de Aminoácidos , Cristalografía por Rayos X , ADN de Plantas/química , Pruebas de Enzimas , Luciferasas/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , Transactivadores/química , Transactivadores/metabolismo
13.
J Immunol ; 198(9): 3627-3636, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28363908

RESUMEN

The cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) mediates innate immune responses against invading pathogens, or against self-dsDNA, which causes autoimmune disorders. Upon nonspecific binding of cytosolic B-form DNA, cGAS synthesizes the second messenger 2'3'-cGAMP and triggers STING-dependent signaling to produce type I IFNs. The cGAS comprises less-conserved N-terminal residues and highly conserved nucleotidyltransferase/Mab21 domains. The function and structure of the well-conserved domains have been extensively studied, whereas the physiological function of the N-terminal domain of cGAS is largely uncharacterized. In this study we used a single-molecule technique combined with traditional biochemical and cellular assays to demonstrate that binding of nonspecific dsDNA by the N-terminal domain of cGAS promotes its activation. We have observed that the N terminus of human cGAS (hcGAS-N160) undergoes secondary structural change upon dsDNA binding in solution. Furthermore, we showed that the hcGAS-N160 helps full length hcGAS to expand the binding range on λDNA and facilitates its binding efficiency to dsDNA compared with hcGAS without the 160 N-terminal residues (hcGAS-d160). More importantly, hcGAS-N160 endows full length hcGAS relatively higher enzyme activity and stronger activation of STING/IRF3-mediated cytosolic DNA signaling. These findings strongly indicate that the N-terminal domain of cGAS plays an important role in enhancing its function.


Asunto(s)
ADN Forma B/metabolismo , Nucleotidiltransferasas/metabolismo , Unión Proteica , Regulación Alostérica , Activación Enzimática , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Dominios Proteicos/genética , Ingeniería de Proteínas , Transducción de Señal
14.
Sci Rep ; 6: 30170, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27460773

RESUMEN

In this work, we developed a method to systematically study the sequence preference of mRNAs during translation initiation. Traditionally, the dynamic process of translation initiation has been studied at the single molecule level with limited sequencing possibility. Using deep sequencing techniques, we identified the sequence preference at different stages of the initiation complexes. Our results provide a comprehensive and dynamic view of the initiation elements in the translation initiation region (TIR), including the S1 binding sequence, the Shine-Dalgarno (SD)/anti-SD interaction and the second codon, at the equilibrium of different initiation complexes. Moreover, our experiments reveal the conformational changes and regional dynamics throughout the dynamic process of mRNA recruitment.


Asunto(s)
Iniciación de la Cadena Peptídica Traduccional/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Secuencia de Bases , Codón Iniciador/genética , Escherichia coli/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos
15.
Sci Rep ; 6: 29119, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27385050

RESUMEN

DNA methylation on CpG sites is the most common epigenetic modification. Recently, methylation in a non-CpG context was found to occur widely on genomic DNA. Moreover, methylation of non-CpG sites is a highly controlled process, and its level may vary during cellular development. To study non-CpG methylation effects on DNA/protein interactions, we have chosen three human transcription factors (TFs): glucocorticoid receptor (GR), brain and muscle ARNT-like 1 (BMAL1) - circadian locomotor output cycles kaput (CLOCK) and estrogen receptor (ER) with methylated or unmethylated DNA binding sequences, using single-molecule and isothermal titration calorimetry assays. The results demonstrated that these TFs interact with methylated DNA with different effects compared with their cognate DNA sequences. The effects of non-CpG methylation on transcriptional regulation were validated by cell-based luciferase assay at protein level. The mechanisms of non-CpG methylation influencing DNA-protein interactions were investigated by crystallographic analyses and molecular dynamics simulation. With BisChIP-seq assays in HEK-293T cells, we found that GR can recognize highly methylated sites within chromatin in cells. Therefore, we conclude that non-CpG methylation of DNA can provide a mechanism for regulating gene expression through directly affecting the binding of TFs.


Asunto(s)
Citosina/metabolismo , Metilación de ADN/genética , Factores de Transcripción/metabolismo , Secuencia de Bases , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Proteínas CLOCK/metabolismo , Inmunoprecipitación de Cromatina , Islas de CpG , Cristalografía por Rayos X , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Recién Nacido , Luciferasas/metabolismo , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Regiones Promotoras Genéticas/genética , Unión Proteica , Dominios Proteicos , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA