Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 627(8003): 407-415, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38383779

RESUMO

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Assuntos
Aquaporina 4 , Autoanticorpos , Autoantígenos , Linfócitos B , Tolerância Imunológica , Neuromielite Óptica , Animais , Humanos , Camundongos , 60533 , Aquaporina 4/deficiência , Aquaporina 4/genética , Aquaporina 4/imunologia , Aquaporina 4/metabolismo , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD40/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , Transcriptoma
2.
Nat Struct Mol Biol ; 30(5): 640-649, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37106137

RESUMO

The Swi2/Snf2 family transcription regulator Modifier of Transcription 1 (Mot1) uses adenosine triphosphate (ATP) to dissociate and reallocate the TATA box-binding protein (TBP) from and between promoters. To reveal how Mot1 removes TBP from TATA box DNA, we determined cryogenic electron microscopy structures that capture different states of the remodeling reaction. The resulting molecular video reveals how Mot1 dissociates TBP in a process that, intriguingly, does not require DNA groove tracking. Instead, the motor grips DNA in the presence of ATP and swings back after ATP hydrolysis, moving TBP to a thermodynamically less stable position on DNA. Dislodged TBP is trapped by a chaperone element that blocks TBP's DNA binding site. Our results show how Swi2/Snf2 proteins can remodel protein-DNA complexes through DNA bending without processive DNA tracking and reveal mechanistic similarities to RNA gripping DEAD box helicases and RIG-I-like immune sensors.


Assuntos
Proteínas de Saccharomyces cerevisiae , Fatores Associados à Proteína de Ligação a TATA , Adenosina Trifosfatases/metabolismo , Fatores de Transcrição/metabolismo , TATA Box , Proteína de Ligação a TATA-Box/química , Proteínas de Saccharomyces cerevisiae/metabolismo , DNA/química , Trifosfato de Adenosina/metabolismo , Fatores Associados à Proteína de Ligação a TATA/química
3.
Mol Cell ; 83(2): 167-185.e9, 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36577401

RESUMO

The DNA double-strand break repair complex Mre11-Rad50-Nbs1 (MRN) detects and nucleolytically processes DNA ends, activates the ATM kinase, and tethers DNA at break sites. How MRN can act both as nuclease and scaffold protein is not well understood. The cryo-EM structure of MRN from Chaetomium thermophilum reveals a 2:2:1 complex with a single Nbs1 wrapping around the autoinhibited Mre11 nuclease dimer. MRN has two DNA-binding modes, one ATP-dependent mode for loading onto DNA ends and one ATP-independent mode through Mre11's C terminus, suggesting how it may interact with DSBs and intact DNA. MRNs two 60-nm-long coiled-coil domains form a linear rod structure, the apex of which is assembled by the two joined zinc-hook motifs. Apices from two MRN complexes can further dimerize, forming 120-nm spanning MRN-MRN structures. Our results illustrate the architecture of MRN and suggest how it mechanistically integrates catalytic and tethering functions.


Assuntos
Reparo do DNA , DNA , Microscopia Crioeletrônica , DNA/genética , Hidrolases Anidrido Ácido/genética , Quebras de DNA de Cadeia Dupla , Enzimas Reparadoras do DNA/metabolismo , Trifosfato de Adenosina/metabolismo , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/metabolismo , Proteínas de Ciclo Celular/metabolismo
4.
Nat Commun ; 13(1): 5464, 2022 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-36115853

RESUMO

Schlafen 11 (SLFN11) is an interferon-inducible antiviral restriction factor with tRNA endoribonuclease and DNA binding functions. It is recruited to stalled replication forks in response to replication stress and inhibits replication of certain viruses such as the human immunodeficiency virus 1 (HIV-1) by modulating the tRNA pool. SLFN11 has been identified as a predictive biomarker in cancer, as its expression correlates with a beneficial response to DNA damage inducing anticancer drugs. However, the mechanism and interdependence of these two functions are largely unknown. Here, we present cryo-electron microscopy (cryo-EM) structures of human SLFN11 in its dimeric apoenzyme state, bound to tRNA and in complex with single-strand DNA. Full-length SLFN11 neither hydrolyses nor binds ATP and the helicase domain appears in an autoinhibited state. Together with biochemical and structure guided mutagenesis studies, our data give detailed insights into the mechanism of endoribonuclease activity as well as suggestions on how SLFN11 may block stressed replication forks.


Assuntos
Antineoplásicos , Antivirais , Microscopia Crioeletrônica , Endorribonucleases , Proteínas Nucleares , Trifosfato de Adenosina , Antineoplásicos/metabolismo , Antivirais/metabolismo , Apoenzimas , DNA , Endorribonucleases/genética , Endorribonucleases/metabolismo , Humanos , Interferons , Proteínas Nucleares/metabolismo , RNA de Transferência/metabolismo
5.
Sci Rep ; 12(1): 15330, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36097274

RESUMO

While recent technological developments contributed to breakthrough advances in single particle cryo-electron microscopy (cryo-EM), sample preparation remains a significant bottleneck for the structure determination of macromolecular complexes. A critical time factor is sample optimization that requires the use of an electron microscope to screen grids prepared under different conditions to achieve the ideal vitreous ice thickness containing the particles. Evaluating sample quality requires access to cryo-electron microscopes and a strong expertise in EM. To facilitate and accelerate the selection procedure of probes suitable for high-resolution cryo-EM, we devised a method to assess the vitreous ice layer thickness of sample coated grids. The experimental setup comprises an optical interferometric microscope equipped with a cryogenic stage and image analysis software based on artificial neural networks (ANN) for an unbiased sample selection. We present and validate this approach for different protein complexes and grid types, and demonstrate its performance for the assessment of ice quality. This technique is moderate in cost and can be easily performed on a laboratory bench. We expect that its throughput and its versatility will contribute to facilitate the sample optimization process for structural biologists.


Assuntos
Gelo , Interferometria , Microscopia Crioeletrônica/métodos , Substâncias Macromoleculares/química , Manejo de Espécimes/métodos
6.
Mol Cell ; 82(18): 3513-3522.e6, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35987200

RESUMO

DNA double-strand breaks (DSBs) threaten genome stability and are linked to tumorigenesis in humans. Repair of DSBs requires the removal of attached proteins and hairpins through a poorly understood but physiologically critical endonuclease activity by the Mre11-Rad50 complex. Here, we report cryoelectron microscopy (cryo-EM) structures of the bacterial Mre11-Rad50 homolog SbcCD bound to a protein-blocked DNA end and a DNA hairpin. The structures reveal that Mre11-Rad50 bends internal DNA for endonucleolytic cleavage and show how internal DNA, DNA ends, and hairpins are processed through a similar ATP-regulated conformational state. Furthermore, Mre11-Rad50 is loaded onto blocked DNA ends with Mre11 pointing away from the block, explaining the distinct biochemistries of 3' → 5' exonucleolytic and endonucleolytic incision through the way Mre11-Rad50 interacts with diverse DNA ends. In summary, our results unify Mre11-Rad50's enigmatic nuclease diversity within a single structural framework and reveal how blocked DNA ends and hairpins are processed.


Assuntos
Proteínas de Ligação a DNA , DNA , Proteína Homóloga a MRE11/química , Hidrolases Anidrido Ácido/genética , Hidrolases Anidrido Ácido/metabolismo , Trifosfato de Adenosina/metabolismo , Microscopia Crioeletrônica , DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Endonucleases/genética , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Humanos , Conformação de Ácido Nucleico
7.
Nucleic Acids Res ; 50(2): 1147-1161, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35037067

RESUMO

The Schlafen family belongs to the interferon-stimulated genes and its members are involved in cell cycle regulation, T cell quiescence, inhibition of viral replication, DNA-repair and tRNA processing. Here, we present the cryo-EM structure of full-length human Schlafen 5 (SLFN5) and the high-resolution crystal structure of the highly conserved N-terminal core domain. We show that the core domain does not resemble an ATPase-like fold and neither binds nor hydrolyzes ATP. SLFN5 binds tRNA as well as single- and double-stranded DNA, suggesting a potential role in transcriptional regulation. Unlike rat Slfn13 or human SLFN11, human SLFN5 did not cleave tRNA. Based on the structure, we identified two residues in proximity to the zinc finger motif that decreased DNA binding when mutated. These results indicate that Schlafen proteins have divergent enzymatic functions and provide a structural platform for future biochemical and genetic studies.


Assuntos
Proteínas de Ciclo Celular/metabolismo , DNA/metabolismo , Humanos , Ligação Proteica , Domínios Proteicos , Transcrição Gênica
8.
Nucleic Acids Res ; 49(17): 10166-10177, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34432045

RESUMO

The cyclic dinucleotide second messenger c-di-AMP is a major player in regulation of potassium homeostasis and osmolyte transport in a variety of bacteria. Along with various direct interactions with proteins such as potassium channels, the second messenger also specifically binds to transcription factors, thereby altering the processes in the cell on the transcriptional level. We here describe the structural and biochemical characterization of BusR from the human pathogen Streptococcus agalactiae. BusR is a member of a yet structurally uncharacterized subfamily of the GntR family of transcription factors that downregulates transcription of the genes for the BusA (OpuA) glycine-betaine transporter upon c-di-AMP binding. We report crystal structures of full-length BusR, its apo and c-di-AMP bound effector domain, as well as cryo-EM structures of BusR bound to its operator DNA. Our structural data, supported by biochemical and biophysical data, reveal that BusR utilizes a unique domain assembly with a tetrameric coiled-coil in between the binding platforms, serving as a molecular ruler to specifically recognize a 22 bp separated bipartite binding motif. Binding of c-di-AMP to BusR induces a shift in equilibrium from an inactivated towards an activated state that allows BusR to bind the target DNA, leading to transcriptional repression.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , DNA Bacteriano/genética , Fosfatos de Dinucleosídeos/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Sistemas do Segundo Mensageiro/genética , Streptococcus agalactiae/genética , Transporte Biológico/genética , Cristalografia por Raios X , Proteínas de Ligação a DNA/genética , Potássio/metabolismo , Domínios Proteicos/genética , Fatores de Transcrição/genética
9.
Nature ; 587(7835): 678-682, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32911480

RESUMO

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.


Assuntos
Domínio Catalítico , Cromatina/química , Cromatina/metabolismo , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/química , Sequência de Aminoácidos , Animais , Autoantígenos/química , Autoantígenos/imunologia , Autoantígenos/metabolismo , Autoantígenos/ultraestrutura , Sítios de Ligação , Ligação Competitiva , Cromatina/genética , Cromatina/ultraestrutura , Microscopia Crioeletrônica , DNA/química , DNA/imunologia , DNA/metabolismo , DNA/ultraestrutura , Ativação Enzimática , Histonas/química , Histonas/metabolismo , Histonas/ultraestrutura , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imunidade Inata , Camundongos , Modelos Moleculares , Nucleossomos/química , Nucleossomos/genética , Nucleossomos/metabolismo , Nucleossomos/ultraestrutura , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/ultraestrutura , Multimerização Proteica , Células THP-1
10.
Structure ; 28(1): 83-95.e5, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31740028

RESUMO

Tel1 (ATM in humans) is a large kinase that resides in the cell in an autoinhibited dimeric state and upon activation orchestrates the cellular response to DNA damage. We report the structure of an endogenous Tel1 dimer from Chaetomium thermophilum. Major parts are at 2.8 Å resolution, including the kinase active site with ATPγS bound, and two different N-terminal solenoid conformations are at 3.4 Å and 3.6 Å, providing a side-chain model for 90% of the Tel1 polypeptide. We show that the N-terminal solenoid has DNA binding activity, but that its movements are not coupled to kinase activation. Although ATPγS and catalytic residues are poised for catalysis, the kinase resides in an autoinhibited state. The PIKK regulatory domain acts as a pseudo-substrate, blocking direct access to the site of catalysis. The structure allows mapping of human cancer mutations and defines mechanisms of autoinhibition at near-atomic resolution.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Proteínas Mutadas de Ataxia Telangiectasia/química , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Chaetomium/enzimologia , Trifosfato de Adenosina/metabolismo , Domínio Catalítico , Chaetomium/química , DNA/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Homeostase , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Multimerização Proteica
11.
Cell Rep ; 29(4): 873-888.e10, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31644910

RESUMO

The CARMA1/CARD11-BCL10-MALT1 (CBM) complex bridges T and B cell antigen receptor (TCR/BCR) ligation to MALT1 protease activation and canonical nuclear factor κB (NF-κB) signaling. Using unbiased mass spectrometry, we discover multiple serine phosphorylation sites in the MALT1 C terminus after T cell activation. Phospho-specific antibodies reveal that CBM-associated MALT1 is transiently hyper-phosphorylated upon TCR/CD28 co-stimulation. We identify a dual role for CK1α as a kinase that is essential for CBM signalosome assembly as well as MALT1 phosphorylation. Although MALT1 phosphorylation is largely dispensable for protease activity, it fosters canonical NF-κB signaling in Jurkat and murine CD4 T cells. Moreover, constitutive MALT1 phosphorylation promotes survival of activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) cells addicted to chronic BCR signaling. Thus, MALT1 phosphorylation triggers optimal NF-κB activation in lymphocytes and survival of lymphoma cells.


Assuntos
Ativação Linfocitária , Linfoma Difuso de Grandes Células B/imunologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Motivos de Aminoácidos , Animais , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Antígenos CD28/metabolismo , Caseína Quinase Ialfa/metabolismo , Células Cultivadas , Guanilato Ciclase/metabolismo , Células HEK293 , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/química , NF-kappa B/metabolismo , Fosforilação
12.
Mol Cell ; 76(3): 382-394.e6, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31492634

RESUMO

DNA double-strand breaks (DSBs) threaten genome stability throughout life and are linked to tumorigenesis in humans. To initiate DSB repair by end joining or homologous recombination, the Mre11-nuclease Rad50-ATPase complex detects and processes diverse and obstructed DNA ends, but a structural mechanism is still lacking. Here we report cryo-EM structures of the E. coli Mre11-Rad50 homolog SbcCD in resting and DNA-bound cutting states. In the resting state, Mre11's nuclease is blocked by ATP-Rad50, and the Rad50 coiled coils appear flexible. Upon DNA binding, the two coiled coils zip up into a rod and, together with the Rad50 nucleotide-binding domains, form a clamp around dsDNA. Mre11 moves to the side of Rad50, binds the DNA end, and assembles a DNA cutting channel for the nuclease reactions. The structures reveal how Mre11-Rad50 can detect and process diverse DNA ends and uncover a clamping and gating function for the coiled coils.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Quebras de DNA de Cadeia Dupla , Replicação do DNA , DNA Bacteriano/metabolismo , Desoxirribonucleases/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Exonucleases/metabolismo , Proteína Homóloga a MRE11/metabolismo , Hidrolases Anidrido Ácido/genética , Hidrolases Anidrido Ácido/ultraestrutura , Microscopia Crioeletrônica , DNA Bacteriano/genética , DNA Bacteriano/ultraestrutura , Desoxirribonucleases/genética , Desoxirribonucleases/ultraestrutura , Escherichia coli/genética , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/ultraestrutura , Exonucleases/genética , Exonucleases/ultraestrutura , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/ultraestrutura , Conformação de Ácido Nucleico , Relação Estrutura-Atividade
14.
Front Immunol ; 9: 2695, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30515170

RESUMO

Assembly of the CARD11/CARMA1-BCL10-MALT1 (CBM) signaling complex upon T or B cell antigen receptor (TCR or BCR) engagement drives lymphocyte activation. Recruitment of pre-assembled BCL10-MALT1 complexes to CARD11 fosters activation of the MALT1 protease and canonical NF-κB signaling. Structural data and in vitro assays have suggested that CARD11 acts as a seed that nucleates the assembly of BCL10 filaments, but the relevance of these findings for CBM complex assembly in cells remains unresolved. To uncouple cellular CARD11 recruitment of BCL10 and BCL10 filament assembly, we generated a BCL10-CARD11 fusion protein that links the C-terminus of BCL10 to the N-terminus of CARD11. When stably expressed in CARD11 KO Jurkat T cells, the BCL10-CARD11 fusion induced constitutive MALT1 activation. Furthermore, in CARD11 KO BJAB B cells, BCL10-CARD11 promoted constitutive NF-κB activation to a similar extent as CARD11 containing oncogenic driver mutations. Using structure-guided destructive mutations in the CARD11-BCL10 (CARD11 R35A) or BCL10-BCL10 (BCL10 R42E) interfaces, we demonstrate that chronic activation by the BCL10-CARD11 fusion protein was independent of the CARD11 CARD. However, activation strictly relied upon the ability of the BCL10 CARD to form oligomers. Thus, by combining distinct CARD mutations in the context of constitutively active BCL10-CARD11 fusion proteins, we provide evidence that BCL10-MALT1 recruitment to CARD11 and BCL10 oligomerization are interconnected processes, which bridge the CARD11 seed to downstream pathways in lymphocytes.


Assuntos
Proteína 10 de Linfoma CCL de Células B/imunologia , Proteínas Adaptadoras de Sinalização CARD/imunologia , Guanilato Ciclase/imunologia , Ativação Linfocitária , Multimerização Proteica/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Proteína 10 de Linfoma CCL de Células B/genética , Proteínas Adaptadoras de Sinalização CARD/genética , Guanilato Ciclase/genética , Células HEK293 , Humanos , Células Jurkat , Multimerização Proteica/genética , Proteínas Recombinantes de Fusão/genética , Linfócitos T/citologia
15.
Nat Commun ; 9(1): 4041, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30279415

RESUMO

The CARD11-BCL10-MALT1 (CBM) complex triggers the adaptive immune response in lymphocytes and lymphoma cells. CARD11/CARMA1 acts as a molecular seed inducing BCL10 filaments, but the integration of MALT1 and the assembly of a functional CBM complex has remained elusive. Using cryo-EM we solved the helical structure of the BCL10-MALT1 filament. The structural model of the filament core solved at 4.9 Å resolution identified the interface between the N-terminal MALT1 DD and the BCL10 caspase recruitment domain. The C-terminal MALT1 Ig and paracaspase domains protrude from this core to orchestrate binding of mediators and substrates at the filament periphery. Mutagenesis studies support the importance of the identified BCL10-MALT1 interface for CBM complex assembly, MALT1 protease activation and NF-κB signaling in Jurkat and primary CD4 T-cells. Collectively, we present a model for the assembly and architecture of the CBM signaling complex and how it functions as a signaling hub in T-lymphocytes.


Assuntos
Proteína 10 de Linfoma CCL de Células B/ultraestrutura , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/ultraestrutura , Proteína 10 de Linfoma CCL de Células B/química , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Microscopia Crioeletrônica , Guanilato Ciclase/metabolismo , Ativação Linfocitária , Modelos Químicos , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/química , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Conformação Proteica
16.
Nucleic Acids Res ; 46(21): 11303-11314, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30277537

RESUMO

The Mre11-Rad50 complex is a DNA double-strand break sensor that cleaves blocked DNA ends and hairpins by an ATP-dependent endo/exonuclease activity for subsequent repair. For that, Mre11-Rad50 complexes, including the Escherichia coli homolog SbcCD, can endonucleolytically cleave one or both strands near a protein block and process free DNA ends via a 3'-5' exonuclease, but a unified basis for these distinct activities is lacking. Here we analyzed DNA binding, ATPase and nuclease reactions on different DNA substrates. SbcCD clips terminal bases of both strands of the DNA end in the presence of ATPγS. It introduces a DNA double-strand break around 20-25 bp from a blocked end after multiple rounds of ATP hydrolysis in a reaction that correlates with local DNA meltability. Interestingly, we find that nuclease reactions on opposing strands are chemically distinct, leaving a 5' phosphate on one strand, but a 3' phosphate on the other strand. Collectively, our results identify an unexpected chemical variability of the nuclease, indicating that the complex is oriented at a free DNA end and facing a block with opposite polarity. This suggests a unified model for ATP-dependent endo- and exonuclease reactions at internal DNA near a block and at free DNA ends.


Assuntos
DNA/metabolismo , Desoxirribonucleases/metabolismo , Proteínas de Escherichia coli/metabolismo , Exonucleases/metabolismo , Trifosfato de Adenosina/metabolismo , DNA/química , Desoxirribonucleases/química , Desoxirribonucleases/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Exonucleases/química , Exonucleases/genética , Polarização de Fluorescência , Hidrólise , Fosfatos/química , Fosfatos/metabolismo , Multimerização Proteica
17.
Elife ; 72018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30047865

RESUMO

The innate immune sensor retinoic acid-inducible gene I (RIG-I) detects cytosolic viral RNA and requires a conformational change caused by both ATP and RNA binding to induce an active signaling state and to trigger an immune response. Previously, we showed that ATP hydrolysis removes RIG-I from lower-affinity self-RNAs (Lässig et al., 2015), revealing how ATP turnover helps RIG-I distinguish viral from self-RNA and explaining why a mutation in a motif that slows down ATP hydrolysis causes the autoimmune disease Singleton-Merten syndrome (SMS). Here we show that a different, mechanistically unexplained SMS variant, C268F, which is localized in the ATP-binding P-loop, can signal independently of ATP but is still dependent on RNA. The structure of RIG-I C268F in complex with double-stranded RNA reveals that C268F helps induce a structural conformation in RIG-I that is similar to that induced by ATP. Our results uncover an unexpected mechanism to explain how a mutation in a P-loop ATPase can induce a gain-of-function ATP state in the absence of ATP.


Assuntos
Adenosina Trifosfatases/química , Trifosfato de Adenosina/química , Doenças da Aorta/genética , Proteína DEAD-box 58/química , Hipoplasia do Esmalte Dentário/genética , Metacarpo/anormalidades , Doenças Musculares/genética , Odontodisplasia/genética , Osteoporose/genética , Calcificação Vascular/genética , Proteínas AAA/química , Proteínas AAA/genética , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/metabolismo , Doenças da Aorta/enzimologia , Doenças da Aorta/patologia , Citosol/virologia , Proteína DEAD-box 58/genética , Hipoplasia do Esmalte Dentário/enzimologia , Hipoplasia do Esmalte Dentário/patologia , Humanos , Hidrólise , Imunidade Inata/genética , Metacarpo/enzimologia , Metacarpo/patologia , Doenças Musculares/enzimologia , Doenças Musculares/patologia , Mutação , Odontodisplasia/enzimologia , Odontodisplasia/patologia , Osteoporose/enzimologia , Osteoporose/patologia , Ligação Proteica , Conformação Proteica , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , RNA Viral/química , RNA Viral/genética , Receptores Imunológicos , Calcificação Vascular/enzimologia , Calcificação Vascular/patologia
18.
Nucleic Acids Res ; 45(20): 12025-12038, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149348

RESUMO

The HerA-NurA helicase-nuclease complex cooperates with Mre11 and Rad50 to coordinate the repair of double-stranded DNA breaks. Little is known, however, about the assembly mechanism and activation of the HerA-NurA. By combining hybrid mass spectrometry with cryo-EM, computational and biochemical data, we investigate the oligomeric formation of HerA and detail the mechanism of nucleotide binding to the HerA-NurA complex from thermophilic archaea. We reveal that ATP-free HerA and HerA-DNA complexes predominantly exist in solution as a heptamer and act as a DNA loading intermediate. The binding of either NurA or ATP stabilizes the hexameric HerA, indicating that HerA-NurA is activated by substrates and complex assembly. To examine the role of ATP in DNA translocation and processing, we investigated how nucleotides interact with the HerA-NurA. We show that while the hexameric HerA binds six nucleotides in an 'all-or-none' fashion, HerA-NurA harbors a highly coordinated pairwise binding mechanism and enables the translocation and processing of double-stranded DNA. Using molecular dynamics simulations, we reveal novel inter-residue interactions between the external ATP and the internal DNA binding sites. Overall, here we propose a stepwise assembly mechanism detailing the synergistic activation of HerA-NurA by ATP, which allows efficient processing of double-stranded DNA.


Assuntos
Proteínas Arqueais/metabolismo , DNA Helicases/metabolismo , DNA Arqueal/metabolismo , Desoxirribonucleases/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Proteínas Arqueais/química , Proteínas Arqueais/genética , Sítios de Ligação/genética , Quebras de DNA de Cadeia Dupla , DNA Helicases/química , DNA Helicases/genética , Reparo do DNA , DNA Arqueal/química , DNA Arqueal/genética , Desoxirribonucleases/química , Desoxirribonucleases/genética , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Sulfolobus solfataricus/enzimologia , Sulfolobus solfataricus/genética , Sulfolobus solfataricus/metabolismo
19.
Eur J Immunol ; 47(3): 458-469, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28058717

RESUMO

Neuromyelitis optica (NMO) is an autoimmune disorder of the central nervous system (CNS) mediated by antibodies to the water channel protein AQP4 expressed in astrocytes. The contribution of AQP4-specific T cells to the class switch recombination of pathogenic AQP4-specific antibodies and the inflammation of the blood-brain barrier is incompletely understood, as immunogenic naturally processed T-cell epitopes of AQP4 are unknown. By immunizing Aqp4-/- mice with full-length murine AQP4 protein followed by recall with overlapping peptides, we here identify AQP4(201-220) as the major immunogenic IAb -restricted epitope of AQP4. We show that WT mice do not harbor AQP4(201-220)-specific T-cell clones in their natural repertoire due to deletional tolerance. However, immunization with AQP4(201-220) of Rag1-/- mice reconstituted with the mature T-cell repertoire of Aqp4-/- mice elicits an encephalomyelitic syndrome. Similarly to the T-cell repertoire, the B-cell repertoire of WT mice is "purged" of AQP4-specific B cells, and robust serum responses to AQP4 are only mounted in Aqp4-/- mice. While AQP4(201-220)-specific T cells alone induce encephalomyelitis, NMO-specific lesional patterns in the CNS and the retina only occur in the additional presence of anti-AQP4 antibodies. Thus, failure of deletional T-cell and B-cell tolerance against AQP4 is a prerequisite for clinically manifest NMO.


Assuntos
Aquaporina 4/metabolismo , Astrócitos/metabolismo , Linfócitos B/fisiologia , Epitopos Imunodominantes/metabolismo , Neuromielite Óptica/imunologia , Retina/imunologia , Linfócitos T/imunologia , Animais , Aquaporina 4/genética , Aquaporina 4/imunologia , Autoanticorpos/metabolismo , Autoimunidade , Deleção Clonal/genética , Células Clonais , Mapeamento de Epitopos , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Switching de Imunoglobulina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Tolerância a Antígenos Próprios
20.
EMBO J ; 35(7): 759-72, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26896444

RESUMO

The Mre11-Rad50-Nbs1 (MRN) complex is a central factor in the repair of DNA double-strand breaks (DSBs). The ATP-dependent mechanisms of how MRN detects and endonucleolytically processes DNA ends for the repair by microhomology-mediated end-joining or further resection in homologous recombination are still unclear. Here, we report the crystal structures of the ATPγS-bound dimer of the Rad50(NBD)(nucleotide-binding domain) from the thermophilic eukaryote Chaetomium thermophilum(Ct) in complex with either DNA or CtMre11(RBD)(Rad50-binding domain) along with small-angle X-ray scattering and cross-linking studies. The structure and DNA binding motifs were validated by DNA binding experiments in vitro and mutational analyses in Saccharomyces cerevisiae in vivo Our analyses provide a structural framework for the architecture of the eukaryotic Mre11-Rad50 complex. They show that a Rad50 dimer binds approximately 18 base pairs of DNA along the dimer interface in anATP-dependent fashion or bridges two DNA ends with a preference for 3' overhangs. Finally, our results may provide a general framework for the interaction of ABC ATPase domains of the Rad50/SMC/RecN protein family with DNA.


Assuntos
Trifosfato de Adenosina/metabolismo , Chaetomium/metabolismo , DNA Fúngico/metabolismo , Proteínas Fúngicas/metabolismo , Chaetomium/genética , Reparo do DNA , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...