Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 184(23): 5728-5739.e16, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34644530

RESUMO

The cyclic pyrimidines 3',5'-cyclic cytidine monophosphate (cCMP) and 3',5'-cyclic uridine monophosphate (cUMP) have been reported in multiple organisms and cell types. As opposed to the cyclic nucleotides 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP), which are second messenger molecules with well-established regulatory roles across all domains of life, the biological role of cyclic pyrimidines has remained unclear. Here we report that cCMP and cUMP are second messengers functioning in bacterial immunity against viruses. We discovered a family of bacterial pyrimidine cyclase enzymes that specifically synthesize cCMP and cUMP following phage infection and demonstrate that these molecules activate immune effectors that execute an antiviral response. A crystal structure of a uridylate cyclase enzyme from this family explains the molecular mechanism of selectivity for pyrimidines as cyclization substrates. Defense systems encoding pyrimidine cyclases, denoted here Pycsar (pyrimidine cyclase system for antiphage resistance), are widespread in prokaryotes. Our results assign clear biological function to cCMP and cUMP as immunity signaling molecules in bacteria.


Assuntos
Bactérias/imunologia , Bactérias/virologia , Bacteriófagos/fisiologia , CMP Cíclico/metabolismo , Nucleotídeos Cíclicos/metabolismo , Uridina Monofosfato/metabolismo , Sequência de Aminoácidos , Bactérias/genética , Burkholderia/enzimologia , CMP Cíclico/química , Ciclização , Escherichia coli/enzimologia , Modelos Moleculares , Mutação/genética , Nucleotídeos Cíclicos/química , Fósforo-Oxigênio Liases/química , Fósforo-Oxigênio Liases/metabolismo , Pirimidinas/metabolismo , Uridina Monofosfato/química
2.
Cell ; 182(1): 38-49.e17, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32544385

RESUMO

cGAS/DncV-like nucleotidyltransferase (CD-NTase) enzymes are immune sensors that synthesize nucleotide second messengers and initiate antiviral responses in bacterial and animal cells. Here, we discover Enterobacter cloacae CD-NTase-associated protein 4 (Cap4) as a founding member of a diverse family of >2,000 bacterial receptors that respond to CD-NTase signals. Structures of Cap4 reveal a promiscuous DNA endonuclease domain activated through ligand-induced oligomerization. Oligonucleotide recognition occurs through an appended SAVED domain that is an unexpected fusion of two CRISPR-associated Rossman fold (CARF) subunits co-opted from type III CRISPR immunity. Like a lock and key, SAVED effectors exquisitely discriminate 2'-5'- and 3'-5'-linked bacterial cyclic oligonucleotide signals and enable specific recognition of at least 180 potential nucleotide second messenger species. Our results reveal SAVED CARF family proteins as major nucleotide second messenger receptors in CBASS and CRISPR immune defense and extend the importance of linkage specificity beyond mammalian cGAS-STING signaling.


Assuntos
Bactérias/virologia , Bacteriófagos/metabolismo , Sistemas CRISPR-Cas , Imunidade , Oligonucleotídeos/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Desoxirribonuclease I/metabolismo , Ligantes , Mutagênese/genética , Nucleotidiltransferases/metabolismo , Ligação Proteica , Sistemas do Segundo Mensageiro
3.
Nature ; 608(7924): 803-807, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35859168

RESUMO

Stimulator of interferon genes (STING) is an antiviral signalling protein that is broadly conserved in both innate immunity in animals and phage defence in prokaryotes1-4. Activation of STING requires its assembly into an oligomeric filament structure through binding of a cyclic dinucleotide4-13, but the molecular basis of STING filament assembly and extension remains unknown. Here we use cryogenic electron microscopy to determine the structure of the active Toll/interleukin-1 receptor (TIR)-STING filament complex from a Sphingobacterium faecium cyclic-oligonucleotide-based antiphage signalling system (CBASS) defence operon. Bacterial TIR-STING filament formation is driven by STING interfaces that become exposed on high-affinity recognition of the cognate cyclic dinucleotide signal c-di-GMP. Repeating dimeric STING units stack laterally head-to-head through surface interfaces, which are also essential for human STING tetramer formation and downstream immune signalling in mammals5. The active bacterial TIR-STING structure reveals further cross-filament contacts that brace the assembly and coordinate packing of the associated TIR NADase effector domains at the base of the filament to drive NAD+ hydrolysis. STING interface and cross-filament contacts are essential for cell growth arrest in vivo and reveal a stepwise mechanism of activation whereby STING filament assembly is required for subsequent effector activation. Our results define the structural basis of STING filament formation in prokaryotic antiviral signalling.


Assuntos
Proteínas de Bactérias , Microscopia Crioeletrônica , Proteínas de Membrana , Receptores de Interleucina-1 , Sphingobacterium , Receptores Toll-Like , Animais , Antivirais/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Bacteriófagos/imunologia , Fosfatos de Dinucleosídeos/metabolismo , Humanos , Imunidade Inata , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Óperon/genética , Receptores de Interleucina-1/química , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/ultraestrutura , Sphingobacterium/química , Sphingobacterium/genética , Sphingobacterium/ultraestrutura , Sphingobacterium/virologia , Receptores Toll-Like/química , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Receptores Toll-Like/ultraestrutura
4.
Proc Natl Acad Sci U S A ; 121(17): e2315018121, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38625940

RESUMO

Heterotrimeric G proteins can be regulated by posttranslational modifications, including ubiquitylation. KCTD5, a pentameric substrate receptor protein consisting of an N-terminal BTB domain and a C-terminal domain, engages CUL3 to form the central scaffold of a cullin-RING E3 ligase complex (CRL3KCTD5) that ubiquitylates Gßγ and reduces Gßγ protein levels in cells. The cryo-EM structure of a 5:5:5 KCTD5/CUL3NTD/Gß1γ2 assembly reveals a highly dynamic complex with rotations of over 60° between the KCTD5BTB/CUL3NTD and KCTD5CTD/Gßγ moieties of the structure. CRL3KCTD5 engages the E3 ligase ARIH1 to ubiquitylate Gßγ in an E3-E3 superassembly, and extension of the structure to include full-length CUL3 with RBX1 and an ARIH1~ubiquitin conjugate reveals that some conformational states position the ARIH1~ubiquitin thioester bond to within 10 Å of lysine-23 of Gß and likely represent priming complexes. Most previously described CRL/substrate structures have consisted of monovalent complexes and have involved flexible peptide substrates. The structure of the KCTD5/CUL3NTD/Gßγ complex shows that the oligomerization of a substrate receptor can generate a polyvalent E3 ligase complex and that the internal dynamics of the substrate receptor can position a structured target for ubiquitylation in a CRL3 complex.


Assuntos
Proteínas de Transporte , Ubiquitina-Proteína Ligases , Ligação Proteica , Ubiquitinação , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Transporte/metabolismo , Ubiquitina/metabolismo , Proteínas Culina/genética , Proteínas Culina/metabolismo
5.
Nature ; 586(7829): 429-433, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32877915

RESUMO

Stimulator of interferon genes (STING) is a receptor in human cells that senses foreign cyclic dinucleotides that are released during bacterial infection and in endogenous cyclic GMP-AMP signalling during viral infection and anti-tumour immunity1-5. STING shares no structural homology with other known signalling proteins6-9, which has limited attempts at functional analysis and prevented explanation of the origin of cyclic dinucleotide signalling in mammalian innate immunity. Here we reveal functional STING homologues encoded within prokaryotic defence islands, as well as a conserved mechanism of signal activation. Crystal structures of bacterial STING define a minimal homodimeric scaffold that selectively responds to cyclic di-GMP synthesized by a neighbouring cGAS/DncV-like nucleotidyltransferase (CD-NTase) enzyme. Bacterial STING domains couple the recognition of cyclic dinucleotides with the formation of protein filaments to drive oligomerization of TIR effector domains and rapid NAD+ cleavage. We reconstruct the evolutionary events that followed the acquisition of STING into metazoan innate immunity, and determine the structure of a full-length TIR-STING fusion from the Pacific oyster Crassostrea gigas. Comparative structural analysis demonstrates how metazoan-specific additions to the core STING scaffold enabled a switch from direct effector function to regulation of antiviral transcription. Together, our results explain the mechanism of STING-dependent signalling and reveal the conservation of a functional cGAS-STING pathway in prokaryotic defence against bacteriophages.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , GMP Cíclico/análogos & derivados , Evolução Molecular , Proteínas de Membrana , Sistemas do Segundo Mensageiro , Animais , Bactérias/química , Bactérias/virologia , Proteínas de Bactérias/química , Bacteriófagos , Cristalografia por Raios X , GMP Cíclico/metabolismo , Proteínas de Membrana/química , Modelos Moleculares , NAD/metabolismo , Nucleotidiltransferases/metabolismo
6.
Mol Cell ; 59(6): 970-83, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26344097

RESUMO

BRCC36 is a Zn(2+)-dependent deubiquitinating enzyme (DUB) that hydrolyzes lysine-63-linked ubiquitin chains as part of distinct macromolecular complexes that participate in either interferon signaling or DNA-damage recognition. The MPN(+) domain protein BRCC36 associates with pseudo DUB MPN(-) proteins KIAA0157 or Abraxas, which are essential for BRCC36 enzymatic activity. To understand the basis for BRCC36 regulation, we have solved the structure of an active BRCC36-KIAA0157 heterodimer and an inactive BRCC36 homodimer. Structural and functional characterizations show how BRCC36 is switched to an active conformation by contacts with KIAA0157. Higher-order association of BRCC36 and KIAA0157 into a dimer of heterodimers (super dimers) was required for DUB activity and interaction with targeting proteins SHMT2 and RAP80. These data provide an explanation of how an inactive pseudo DUB allosterically activates a cognate DUB partner and implicates super dimerization as a new regulatory mechanism underlying BRCC36 DUB activity, subcellular localization, and biological function.


Assuntos
Formigas/enzimologia , Proteínas de Insetos/química , Proteínas Associadas à Matriz Nuclear/química , Proteases Específicas de Ubiquitina/química , Animais , Domínio Catalítico , Cristalografia por Raios X , Enzimas Desubiquitinantes , Células HEK293 , Células HeLa , Humanos , Proteínas de Insetos/fisiologia , Cinética , Proteínas de Membrana/química , Modelos Moleculares , Proteínas Associadas à Matriz Nuclear/fisiologia , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Proteases Específicas de Ubiquitina/fisiologia
7.
J Am Chem Soc ; 143(34): 13473-13477, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34403584

RESUMO

Employed for over half a century to study protein synthesis, cycloheximide (CHX, 1) is a small molecule natural product that reversibly inhibits translation elongation. More recently, CHX has been applied to ribosome profiling, a method for mapping ribosome positions on mRNA genome-wide. Despite CHX's extensive use, CHX treatment often results in incomplete translation inhibition due to its rapid reversibility, prompting the need for improved reagents. Here, we report the concise synthesis of C13-amide-functionalized CHX derivatives with increased potencies toward protein synthesis inhibition. Cryogenic electron microscopy (cryo-EM) revealed that C13-aminobenzoyl CHX (8) occupies the same site as CHX, competing with the 3' end of E-site tRNA. We demonstrate that 8 is superior to CHX for ribosome profiling experiments, enabling more effective capture of ribosome conformations through sustained stabilization of polysomes. Our studies identify powerful chemical reagents to study protein synthesis and reveal the molecular basis of their enhanced potency.


Assuntos
Produtos Biológicos/farmacologia , Cicloeximida/análogos & derivados , Elongação Traducional da Cadeia Peptídica/efeitos dos fármacos , Amidas/química , Produtos Biológicos/química , Cicloeximida/metabolismo , Cicloeximida/farmacologia , Células HEK293 , Humanos , RNA de Transferência/química , RNA de Transferência/metabolismo , Ribossomos/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(6): 1311-1316, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28115697

RESUMO

IpaH enzymes are secreted bacterial effectors that function within host cells as E3 ubiquitin (Ub) ligases. Catalytic activity is imparted by a conserved novel E3 ligase (NEL) domain that is unique to Gram-negative pathogens and whose activity is repressed by a flanking substrate-binding leucine-rich repeat (LRR) domain when substrate is absent. How the NEL domain catalyzes the conjugation of Ub onto substrates, recognizes host E2s, and maintains its autoinhibited state remain poorly understood. Here we used mutagenesis and enzyme kinetic analyses to address these gaps in knowledge. Mutagenesis of conserved residues on two remote surfaces of the NEL domain identified functional clusters proximal to and distal to the active site cysteine. By analyzing the kinetics of Ub charging and discharging, we identified proximal active site residues that function as either the catalytic acid or catalytic base for aminolysis. Further analysis revealed that distal site residues mediate the direct binding of E2. In studying the full-length protein, we also have uncovered that IpaH family autoinhibition is achieved by a short-circuiting mechanism wherein the LRR domain selectively blocks productive aminolysis, but not the nonproductive discharge of Ub from the E3 to solvent. This mode of autoinhibition, which is not shared by the HECT domain ligase Smurf2, leads to the unanticipated depletion of E2∼Ub and thus a concomitant dominant-negative effect on other E3s in vitro, raising the possibility that short circuiting also may serve to restrict the function of host E3s in cells.


Assuntos
Proteínas de Bactérias/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Bactérias/genética , Catálise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonella/enzimologia , Shigella/enzimologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
9.
Structure ; 32(6): 795-811.e6, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38531363

RESUMO

GCN2 is a stress response kinase that phosphorylates the translation initiation factor eIF2α to inhibit general protein synthesis when activated by uncharged tRNA and stalled ribosomes. The presence of a HisRS-like domain in GCN2, normally associated with tRNA aminoacylation, led to the hypothesis that eIF2α kinase activity is regulated by the direct binding of this domain to uncharged tRNA. Here we solved the structure of the HisRS-like domain in the context of full-length GCN2 by cryoEM. Structure and function analysis shows the HisRS-like domain of GCN2 has lost histidine and ATP binding but retains tRNA binding abilities. Hydrogen deuterium exchange mass spectrometry, site-directed mutagenesis and computational docking experiments support a tRNA binding model that is partially shifted from that employed by bona fide HisRS enzymes. These results demonstrate that the HisRS-like domain of GCN2 is a pseudoenzyme and advance our understanding of GCN2 regulation and function.


Assuntos
Ligação Proteica , Proteínas Serina-Treonina Quinases , RNA de Transferência , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , RNA de Transferência/metabolismo , RNA de Transferência/química , Sítios de Ligação , Domínios Proteicos , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Microscopia Crioeletrônica , Simulação de Acoplamento Molecular , Modelos Moleculares , Trifosfato de Adenosina/metabolismo , Saccharomyces cerevisiae/metabolismo , Humanos , Histidina/metabolismo , Histidina/química , Fosforilação
10.
Commun Biol ; 7(1): 92, 2024 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216676

RESUMO

Acyl carrier protein (ACP) is the work horse of polyketide (PKS) and fatty acid synthases (FAS) and acts as a substrate shuttling domain in these mega enzymes. In fungi, FAS forms a 2.6 MDa symmetric assembly with six identical copies of FAS1 and FAS2 polypeptides. However, ACP spatial distribution is not restricted by symmetry owing to the long and flexible loops that tether the shuttling domain to its corresponding FAS2 polypeptide. This symmetry breaking has hampered experimental investigation of substrate shuttling route in fungal FAS. Here, we develop a protein engineering and expression method to isolate asymmetric fungal FAS proteins containing odd numbers of ACP domains. Electron cryomicroscopy (cryoEM) observation of the engineered complex reveals a non-uniform distribution of the substrate shuttling domain relative to its corresponding FAS2 polypeptide at 2.9 Å resolution. This work lays the methodological foundation for experimental study of ACP shuttling route in fungi.


Assuntos
Proteína de Transporte de Acila , Saccharomyces cerevisiae , Animais , Cavalos , Proteína de Transporte de Acila/química , Saccharomyces cerevisiae/metabolismo , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/química , Proteínas Fúngicas/metabolismo , Peptídeos/metabolismo
11.
J Biol Chem ; 287(1): 268-275, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22065585

RESUMO

The IpaH family of novel E3 ligase (NEL) enzymes occur in a variety of pathogenic and commensal bacteria that interact with eukaryotic hosts. We demonstrate that the leucine-rich repeat (LRR) substrate recognition domains of different IpaH enzymes autoinhibit the enzymatic activity of the adjacent catalytic novel E3 ligase domain by two distinct but conserved structural mechanisms. Autoinhibition is required for the in vivo biological activity of two IpaH enzymes in a eukaryotic model system. Autoinhibition was retro-engineered into a constitutively active IpaH enzyme from Yersinia pestis by introduction of single site substitutions, thereby demonstrating the conservation of autoregulatory infrastructure across the IpaH enzyme family.


Assuntos
Sequência Conservada , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/química , Substituição de Aminoácidos , Modelos Moleculares , Estrutura Terciária de Proteína , Shigella flexneri/enzimologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Yersinia pestis/enzimologia
12.
Nat Commun ; 14(1): 3460, 2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37308485

RESUMO

Fatty acid synthase (FASN) catalyzes the de novo synthesis of palmitate, a 16-carbon chain fatty acid that is the primary precursor of lipid metabolism and an important intracellular signaling molecule. FASN is an attractive drug target in diabetes, cancer, fatty liver diseases, and viral infections. Here, we develop an engineered full-length human FASN (hFASN) that enables isolation of the condensing and modifying regions of the protein post-translation. The engineered protein enables electron cryo-microscopy (cryoEM) structure determination of the core modifying region of hFASN to 2.7 Å resolution. Examination of the dehydratase dimer within this region reveals that unlike its close homolog, porcine FASN, the catalytic cavity is close-ended and is accessible only through one opening in the vicinity of the active site. The core modifying region exhibits two major global conformational variabilities that describe long-range bending and twisting motions of the complex in solution. Finally, we solved the structure of this region bound to an anti-cancer drug, Denifanstat (i.e., TVB-2640), demonstrating the utility of our approach as a platform for structure guided design of future hFASN small molecule inhibitors.


Assuntos
Carbono , Ácido Graxo Sintases , Humanos , Animais , Suínos , Catálise , Microscopia Crioeletrônica , Sistemas de Liberação de Medicamentos
13.
Nat Struct Mol Biol ; 28(12): 1029-1037, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34887561

RESUMO

Close coordination between chaperones is essential for protein biosynthesis, including the delivery of tail-anchored (TA) proteins containing a single C-terminal transmembrane domain to the endoplasmic reticulum (ER) by the conserved GET pathway. For successful targeting, nascent TA proteins must be promptly chaperoned and loaded onto the cytosolic ATPase Get3 through a transfer reaction involving the chaperone SGTA and bridging factors Get4, Ubl4a and Bag6. Here, we report cryo-electron microscopy structures of metazoan pretargeting GET complexes at 3.3-3.6 Å. The structures reveal that Get3 helix 8 and the Get4 C terminus form a composite lid over the Get3 substrate-binding chamber that is opened by SGTA. Another interaction with Get4 prevents formation of Get3 helix 4, which links the substrate chamber and ATPase domain. Both interactions facilitate TA protein transfer from SGTA to Get3. Our findings show how the pretargeting complex primes Get3 for coordinated client loading and ER targeting.


Assuntos
ATPases Transportadoras de Arsenito/metabolismo , Chaperonas Moleculares/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Microscopia Crioeletrônica , Retículo Endoplasmático/metabolismo , Humanos , Modelos Moleculares , Conformação Proteica , Ubiquitinas/metabolismo , Peixe-Zebra
14.
J Mol Biol ; 432(4): 952-966, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31634471

RESUMO

Ubiquitin-conjugating E2 enzymes are central to the ubiquitination cascade and have been implicated in cancer and other diseases. Despite strong interest in developing specific E2 inhibitors, the shallow and exposed active site has proven recalcitrant to targeting with reversible small-molecule inhibitors. Here, we used phage display to generate highly potent and selective ubiquitin variants (UbVs) that target the E2 backside, which is located opposite to the active site. A UbV targeting Ube2D1 did not affect charging but greatly attenuated chain elongation. Likewise, a UbV targeting the E2 variant Ube2V1 did not interfere with the charging of its partner E2 enzyme but inhibited formation of diubiquitin. In contrast, a UbV that bound to the backside of Ube2G1 impeded the generation of thioester-linked ubiquitin to the active site cysteine of Ube2G1 by the E1 enzyme. Crystal structures of UbVs in complex with three E2 proteins revealed distinctive molecular interactions in each case, but they also highlighted a common backside pocket that the UbVs used for enhanced affinity and specificity. These findings validate the E2 backside as a target for inhibition and provide structural insights to aid inhibitor design and screening efforts.


Assuntos
Inibidores Enzimáticos/farmacologia , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina/metabolismo , Sequência de Aminoácidos , Inibidores Enzimáticos/química , Humanos , Biblioteca de Peptídeos , Ligação Proteica , Engenharia de Proteínas/métodos , Estrutura Secundária de Proteína , Enzimas de Conjugação de Ubiquitina/antagonistas & inibidores , Enzimas de Conjugação de Ubiquitina/química , Ubiquitinação
15.
Nat Struct Mol Biol ; 26(5): 343-349, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31011209

RESUMO

Aberrantly stalled ribosomes initiate the ribosome-associated quality control (RQC) and mRNA surveillance pathways for the degradation of potentially toxic peptides and faulty mRNAs. During RQC, ANKZF1 (yeast Vms1p) releases ubiquitinated nascent proteins from 60S ribosomal subunits for proteasomal degradation. Here, we use a cell-free system to show that ANKZF1 and Vms1p sever polypeptidyl-tRNAs on RQC complexes by precisely cleaving off the terminal 3'CCA nucleotides universal to all tRNAs. This produces a tRNA fragment that cannot be aminoacylated until its 3'CCA end is restored. The recycling of ANKZF1-cleaved tRNAs is intact in the mammalian cytosol via a two-step process that requires the removal of a 2',3'-cyclic phosphate and TRNT1, the sole CCA-adding enzyme that mediates tRNA biogenesis in eukaryotes. TRNT1 also discriminates between properly folded tRNA substrates and aberrant tRNA substrates, selectively tagging the latter for degradation. Thus, ANKZF1 liberates peptidyl-tRNAs from stalled ribosomes such that the tRNA is checked in an obligate way for integrity before reentry into the translation cycle.


Assuntos
Proteínas de Transporte/metabolismo , RNA de Transferência/metabolismo , Ribossomos/metabolismo , Células HEK293 , Humanos , Biossíntese de Proteínas , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato
16.
Mol Cell Biol ; 34(3): 362-73, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24248594

RESUMO

IpaH proteins are bacterium-specific E3 enzymes that function as type three secretion system (T3SS) effectors in Salmonella, Shigella, and other Gram-negative bacteria. IpaH enzymes recruit host substrates for ubiquitination via a leucine-rich repeat (LRR) domain, which can inhibit the catalytic domain in the absence of substrate. The basis for substrate recognition and the alleviation of autoinhibition upon substrate binding is unknown. Here, we report the X-ray structure of Salmonella SspH1 in complex with human PKN1. The LRR domain of SspH1 interacts specifically with the HR1b coiled-coil subdomain of PKN1 in a manner that sterically displaces the catalytic domain from the LRR domain, thereby activating catalytic function. SspH1 catalyzes the ubiquitination and proteasome-dependent degradation of PKN1 in cells, which attenuates androgen receptor responsiveness but not NF-κB activity. These regulatory features are conserved in other IpaH-substrate interactions. Our results explain the mechanism whereby substrate recognition and enzyme autoregulation are coupled in this class of bacterial ubiquitin ligases.


Assuntos
Proteínas de Bactérias/metabolismo , Proteína Quinase C/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação/genética , Domínio Catalítico , Cristalografia por Raios X , Células HEK293 , Humanos , Immunoblotting , Modelos Moleculares , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteína Quinase C/química , Proteína Quinase C/genética , Estrutura Terciária de Proteína , Receptores Androgênicos/metabolismo , Salmonella/genética , Salmonella/metabolismo , Especificidade por Substrato , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA