Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
PLoS Biol ; 22(3): e3002567, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38470934

RESUMO

PEX5, the peroxisomal protein shuttling receptor, binds newly synthesized proteins in the cytosol and transports them to the organelle. During its stay at the peroxisomal protein translocon, PEX5 is monoubiquitinated at its cysteine 11 residue, a mandatory modification for its subsequent ATP-dependent extraction back into the cytosol. The reason why a cysteine and not a lysine residue is the ubiquitin acceptor is unknown. Using an established rat liver-based cell-free in vitro system, we found that, in contrast to wild-type PEX5, a PEX5 protein possessing a lysine at position 11 is polyubiquitinated at the peroxisomal membrane, a modification that negatively interferes with the extraction process. Wild-type PEX5 cannot retain a polyubiquitin chain because ubiquitination at cysteine 11 is a reversible reaction, with the E2-mediated deubiquitination step presenting faster kinetics than PEX5 polyubiquitination. We propose that the reversible nonconventional ubiquitination of PEX5 ensures that neither the peroxisomal protein translocon becomes obstructed with polyubiquitinated PEX5 nor is PEX5 targeted for proteasomal degradation.


Assuntos
Cisteína , Lisina , Animais , Ratos , Proteínas de Transporte/metabolismo , Cisteína/metabolismo , Lisina/metabolismo , Receptor 1 de Sinal de Orientação para Peroxissomos/química , Receptor 1 de Sinal de Orientação para Peroxissomos/metabolismo , Transporte Proteico , Ubiquitinação
2.
PLoS Pathog ; 20(3): e1012100, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38527094

RESUMO

The coronavirus papain-like protease (PLpro) is crucial for viral replicase polyprotein processing. Additionally, PLpro can subvert host defense mechanisms by its deubiquitinating (DUB) and deISGylating activities. To elucidate the role of these activities during SARS-CoV-2 infection, we introduced mutations that disrupt binding of PLpro to ubiquitin or ISG15. We identified several mutations that strongly reduced DUB activity of PLpro, without affecting viral polyprotein processing. In contrast, mutations that abrogated deISGylating activity also hampered viral polyprotein processing and when introduced into the virus these mutants were not viable. SARS-CoV-2 mutants exhibiting reduced DUB activity elicited a stronger interferon response in human lung cells. In a mouse model of severe disease, disruption of PLpro DUB activity did not affect lethality, virus replication, or innate immune responses in the lungs. This suggests that the DUB activity of SARS-CoV-2 PLpro is dispensable for virus replication and does not affect innate immune responses in vivo. Interestingly, the DUB mutant of SARS-CoV replicated to slightly lower titers in mice and elicited a diminished immune response early in infection, although lethality was unaffected. We previously showed that a MERS-CoV mutant deficient in DUB and deISGylating activity was strongly attenuated in mice. Here, we demonstrate that the role of PLpro DUB activity during infection can vary considerably between highly pathogenic coronaviruses. Therefore, careful considerations should be taken when developing pan-coronavirus antiviral strategies targeting PLpro.


Assuntos
COVID-19 , Proteases Semelhantes à Papaína de Coronavírus , Humanos , Animais , Camundongos , Proteases Semelhantes à Papaína de Coronavírus/genética , SARS-CoV-2/metabolismo , Imunidade Inata , Papaína/genética , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , Replicação Viral , Poliproteínas
3.
Nat Chem Biol ; 20(2): 190-200, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37620400

RESUMO

Ubiquitin (Ub) chain formation by homologous to E6AP C-terminus (HECT)-family E3 ligases regulates vast biology, yet the structural mechanisms remain unknown. We used chemistry and cryo-electron microscopy (cryo-EM) to visualize stable mimics of the intermediates along K48-linked Ub chain formation by the human E3, UBR5. The structural data reveal a ≈ 620 kDa UBR5 dimer as the functional unit, comprising a scaffold with flexibly tethered Ub-associated (UBA) domains, and elaborately arranged HECT domains. Chains are forged by a UBA domain capturing an acceptor Ub, with its K48 lured into the active site by numerous interactions between the acceptor Ub, manifold UBR5 elements and the donor Ub. The cryo-EM reconstructions allow defining conserved HECT domain conformations catalyzing Ub transfer from E2 to E3 and from E3. Our data show how a full-length E3, ubiquitins to be adjoined, E2 and intermediary products guide a feed-forward HECT domain conformational cycle establishing a highly efficient, broadly targeting, K48-linked Ub chain forging machine.


Assuntos
Ubiquitina-Proteína Ligases , Ubiquitina , Humanos , Ubiquitina/química , Microscopia Crioeletrônica , Ubiquitina-Proteína Ligases/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Ubiquitinação
4.
Nature ; 587(7835): 657-662, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32726803

RESUMO

The papain-like protease PLpro is an essential coronavirus enzyme that is required for processing viral polyproteins to generate a functional replicase complex and enable viral spread1,2. PLpro is also implicated in cleaving proteinaceous post-translational modifications on host proteins as an evasion mechanism against host antiviral immune responses3-5. Here we perform biochemical, structural and functional characterization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PLpro (SCoV2-PLpro) and outline differences with SARS-CoV PLpro (SCoV-PLpro) in regulation of host interferon and NF-κB pathways. SCoV2-PLpro and SCoV-PLpro share 83% sequence identity but exhibit different host substrate preferences; SCoV2-PLpro preferentially cleaves the ubiquitin-like interferon-stimulated gene 15 protein (ISG15), whereas SCoV-PLpro predominantly targets ubiquitin chains. The crystal structure of SCoV2-PLpro in complex with ISG15 reveals distinctive interactions with the amino-terminal ubiquitin-like domain of ISG15, highlighting the high affinity and specificity of these interactions. Furthermore, upon infection, SCoV2-PLpro contributes to the cleavage of ISG15 from interferon responsive factor 3 (IRF3) and attenuates type I interferon responses. Notably, inhibition of SCoV2-PLpro with GRL-0617 impairs the virus-induced cytopathogenic effect, maintains the antiviral interferon pathway and reduces viral replication in infected cells. These results highlight a potential dual therapeutic strategy in which targeting of SCoV2-PLpro can suppress SARS-CoV-2 infection and promote antiviral immunity.


Assuntos
COVID-19/imunologia , COVID-19/virologia , Proteases Semelhantes à Papaína de Coronavírus/química , Proteases Semelhantes à Papaína de Coronavírus/metabolismo , Imunidade Inata , SARS-CoV-2/enzimologia , SARS-CoV-2/imunologia , Animais , Proteases Semelhantes à Papaína de Coronavírus/antagonistas & inibidores , Citocinas/química , Citocinas/metabolismo , Enzimas Desubiquitinantes/antagonistas & inibidores , Enzimas Desubiquitinantes/química , Enzimas Desubiquitinantes/metabolismo , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferons/imunologia , Interferons/metabolismo , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , NF-kappa B/imunologia , NF-kappa B/metabolismo , Ligação Proteica , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/fisiologia , Ubiquitinação , Ubiquitinas/química , Ubiquitinas/metabolismo , Tratamento Farmacológico da COVID-19
5.
Semin Cell Dev Biol ; 132: 74-85, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-34961664

RESUMO

Protein ubiquitination is a key post-translational modification in regulating many fundamental cellular processes and dysregulation of these processes can give rise to a vast array of diseases. Unravelling the molecular mechanisms of ubiquitination hence is an important area in current ubiquitin research with as aim to understand this enigmatic process. The complexity of ubiquitin (Ub) signaling arises from the large variety of Ub conjugates, where Ub is attached to other Ub proteins, Ub-like proteins, and protein substrates. The chemical preparation of such Ub conjugates in high homogeneity and in adequate amounts contributes greatly to the deciphering of Ub signaling. The strength of these chemically synthesized conjugates lies in the chemo-selectivity in which they can be created that are sometimes difficult to obtain using biochemical methodology. In this review, we will discuss the progress in the chemical protein synthesis of state-of-the-art Ub and Ub-like chemical probes, their unique concepts and related discoveries in the ubiquitin field.


Assuntos
Processamento de Proteína Pós-Traducional , Ubiquitina , Ubiquitina/metabolismo , Ubiquitinação , Proteínas/metabolismo , Transdução de Sinais
6.
EMBO J ; 39(18): e106275, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32845033

RESUMO

The SARS-CoV-2 coronavirus encodes an essential papain-like protease domain as part of its non-structural protein (nsp)-3, namely SARS2 PLpro, that cleaves the viral polyprotein, but also removes ubiquitin-like ISG15 protein modifications as well as, with lower activity, Lys48-linked polyubiquitin. Structures of PLpro bound to ubiquitin and ISG15 reveal that the S1 ubiquitin-binding site is responsible for high ISG15 activity, while the S2 binding site provides Lys48 chain specificity and cleavage efficiency. To identify PLpro inhibitors in a repurposing approach, screening of 3,727 unique approved drugs and clinical compounds against SARS2 PLpro identified no compounds that inhibited PLpro consistently or that could be validated in counterscreens. More promisingly, non-covalent small molecule SARS PLpro inhibitors also target SARS2 PLpro, prevent self-processing of nsp3 in cells and display high potency and excellent antiviral activity in a SARS-CoV-2 infection model.


Assuntos
Antivirais/farmacologia , Proteases 3C de Coronavírus/antagonistas & inibidores , Proteases 3C de Coronavírus/metabolismo , SARS-CoV-2/metabolismo , Ubiquitina/metabolismo , Animais , Sítios de Ligação , Chlorocebus aethiops , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/genética , Cristalografia por Raios X , Citocinas/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Reposicionamento de Medicamentos , Polarização de Fluorescência , Células HEK293 , Humanos , Cinética , Modelos Moleculares , Inibidores de Proteases/farmacologia , Conformação Proteica , SARS-CoV-2/química , SARS-CoV-2/genética , Ubiquitinas/genética , Células Vero
7.
Mol Cell ; 62(4): 572-85, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27203180

RESUMO

Deubiquitinating enzymes (DUBs) recognize and cleave linkage-specific polyubiquitin (polyUb) chains, but mechanisms underlying specificity remain elusive in many cases. The severe acute respiratory syndrome (SARS) coronavirus papain-like protease (PLpro) is a DUB that cleaves ISG15, a two-domain Ub-like protein, and Lys48-linked polyUb chains, releasing diUb(Lys48) products. To elucidate this specificity, we report the 2.85 Å crystal structure of SARS PLpro bound to a diUb(Lys48) activity-based probe. SARS PLpro binds diUb(Lys48) in an extended conformation via two contact sites, S1 and S2, which are proximal and distal to the active site, respectively. We show that specificity for polyUb(Lys48) chains is predicated on contacts in the S2 site and enhanced by an S1-S1' preference for a Lys48 linkage across the active site. In contrast, ISG15 specificity is dominated by contacts in the S1 site. Determinants revealed for polyUb(Lys48) specificity should prove useful in understanding PLpro deubiquitinating activities in coronavirus infections.


Assuntos
Cisteína Endopeptidases/metabolismo , Citocinas/metabolismo , Enzimas Desubiquitinantes/metabolismo , Poliubiquitina/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/enzimologia , Ubiquitinas/metabolismo , Proteínas Virais/metabolismo , Sítios de Ligação , Proteases 3C de Coronavírus , Cisteína Endopeptidases/química , Cisteína Endopeptidases/genética , Citocinas/química , Enzimas Desubiquitinantes/química , Células HeLa , Humanos , Lisina , Modelos Moleculares , Mutação , Poliubiquitina/química , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Relação Estrutura-Atividade , Ubiquitinação , Ubiquitinas/química , Proteínas Virais/química , Proteínas Virais/genética
8.
Chembiochem ; 24(2): e202200601, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36377600

RESUMO

Small ubiquitin-like modifiers (SUMOs) are conjugated to protein substrates in cells to regulate their function. The attachment of SUMO family members SUMO1-3 to substrate proteins is reversed by specific isopeptidases called SENPs (sentrin-specific protease). Whereas SENPs are SUMO-isoform or linkage type specific, comprehensive analysis is missing. Furthermore, the underlying mechanism of SENP linkage specificity remains unclear. We present a high-throughput synthesis of 83 isopeptide-linked SUMO-based fluorescence polarization reagents to study enzyme preferences. The assay reagents were synthesized via a native chemical ligation-desulfurization protocol between 11-mer peptides containing a γ-thiolysine and a SUMO3 thioester. Subsequently, five recombinantly expressed SENPs were screened using these assay reagents to reveal their deconjugation activity and substrate preferences. In general, we observed that SENP1 is the most active and nonselective SENP while SENP6 and SENP7 show the least activity. Furthermore, SENPs differentially process peptides derived from SUMO1-3, who form a minimalistic representation of diSUMO chains. To validate our findings, five distinct isopeptide-linked diSUMO chains were chemically synthesized and proteolysis was monitored using a gel-based read-out.


Assuntos
Corantes Fluorescentes , Ensaios de Triagem em Larga Escala , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina , Endopeptidases/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Peptídeo Hidrolases/metabolismo , Peptídeos/química , Proteólise , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/síntese química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química
9.
Nat Chem Biol ; 17(3): 272-279, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33288957

RESUMO

Virtually all aspects of cell biology are regulated by a ubiquitin code where distinct ubiquitin chain architectures guide the binding events and itineraries of modified substrates. Various combinations of E2 and E3 enzymes accomplish chain formation by forging isopeptide bonds between the C terminus of their transiently linked donor ubiquitin and a specific nucleophilic amino acid on the acceptor ubiquitin, yet it is unknown whether the fundamental feature of most acceptors-the lysine side chain-affects catalysis. Here, use of synthetic ubiquitins with non-natural acceptor site replacements reveals that the aliphatic side chain specifying reactive amine geometry is a determinant of the ubiquitin code, through unanticipated and complex reliance of many distinct ubiquitin-carrying enzymes on a canonical acceptor lysine.


Assuntos
Lisina/química , Proteína NEDD8/química , Poliubiquitina/química , Processamento de Proteína Pós-Traducional , Ubiquitina/química , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Cinética , Lisina/metabolismo , Modelos Moleculares , Proteína NEDD8/genética , Proteína NEDD8/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poliubiquitina/genética , Poliubiquitina/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação
10.
J Biol Chem ; 297(2): 100925, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34214498

RESUMO

Apart from prevention using vaccinations, the management options for COVID-19 remain limited. In retrospective cohort studies, use of famotidine, a specific oral H2 receptor antagonist (antihistamine), has been associated with reduced risk of intubation and death in patients hospitalized with COVID-19. In a case series, nonhospitalized patients with COVID-19 experienced rapid symptom resolution after taking famotidine, but the molecular basis of these observations remains elusive. Here we show using biochemical, cellular, and functional assays that famotidine has no effect on viral replication or viral protease activity. However, famotidine can affect histamine-induced signaling processes in infected Caco2 cells. Specifically, famotidine treatment inhibits histamine-induced expression of Toll-like receptor 3 (TLR3) in SARS-CoV-2 infected cells and can reduce TLR3-dependent signaling processes that culminate in activation of IRF3 and the NF-κB pathway, subsequently controlling antiviral and inflammatory responses. SARS-CoV-2-infected cells treated with famotidine demonstrate reduced expression levels of the inflammatory mediators CCL-2 and IL6, drivers of the cytokine release syndrome that precipitates poor outcome for patients with COVID-19. Given that pharmacokinetic studies indicate that famotidine can reach concentrations in blood that suffice to antagonize histamine H2 receptors expressed in mast cells, neutrophils, and eosinophils, these observations explain how famotidine may contribute to the reduced histamine-induced inflammation and cytokine release, thereby improving the outcome for patients with COVID-19.


Assuntos
Famotidina/farmacologia , Antagonistas dos Receptores Histamínicos/farmacologia , SARS-CoV-2/efeitos dos fármacos , Receptor 3 Toll-Like/metabolismo , Células A549 , Sítios de Ligação , Células CACO-2 , Quimiocina CCL2/metabolismo , Proteases 3C de Coronavírus/metabolismo , Células HeLa , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interleucina-6/metabolismo , Simulação de Acoplamento Molecular , NF-kappa B/metabolismo , Ligação Proteica , SARS-CoV-2/fisiologia , Transdução de Sinais , Receptor 3 Toll-Like/química , Replicação Viral
11.
J Am Chem Soc ; 144(45): 20582-20589, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36318515

RESUMO

We describe the development and optimization of a methodology to prepare peptides and proteins modified on the arginine residue with an adenosine-di-phosphate-ribosyl (ADPr) group. Our method comprises reacting an ornithine containing polypeptide on-resin with an α-linked anomeric isothiourea N-riboside, ensuing installment of a phosphomonoester at the 5'-hydroxyl of the ribosyl moiety followed by the conversion into the adenosine diphosphate. We use this method to obtain four regioisomers of ADP-ribosylated ubiquitin (UbADPr), each modified with an ADP-ribosyl residue on a different arginine position within the ubiquitin (Ub) protein (Arg42, Arg54, Arg72, and Arg74) as the first reported examples of fully synthetic arginine-linked ADPr-modified proteins. We show the chemically prepared Arg-linked UbADPr to be accepted and processed by Legionella enzymes and compare the entire suite of four Arg-linked UbADPr regioisomers in a variety of biochemical experiments, allowing us to profile the activity and selectivity of Legionella pneumophila ligase and hydrolase enzymes.


Assuntos
Adenosina Difosfato Ribose , Arginina , Adenosina Difosfato Ribose/química , Arginina/metabolismo , ADP-Ribosilação , Ubiquitina/química , Proteínas Ubiquitinadas/metabolismo , Peptídeos/química
12.
Chembiochem ; 23(19): e202200304, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-35920208

RESUMO

Chemical protein synthesis has proven to be a powerful tool to access homogenously modified proteins. The chemical synthesis of nanobodies (Nb) would create possibilities to design tailored Nbs with a range of chemical modifications such as tags, linkers, reporter groups, and subsequently, Nb-drug conjugates. Herein, we describe the total chemical synthesis of a 123 amino-acid Nb against GFP. A native chemical ligation- desulfurization strategy was successfully applied for the synthesis of this GFP Nb, modified with a propargyl (PA) moiety for on-demand functionalization. Biophysical characterization indicated that the synthetic GFP Nb-PA was correctly folded after internal disulfide bond formation. The synthetic Nb-PA was functionalized with a biotin or a sulfo-cyanine5 dye by copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC), resulting in two distinct probes used for functional in vitro validation in pull-down and confocal microscopy settings.


Assuntos
Azidas , Anticorpos de Domínio Único , Alcinos/química , Azidas/química , Biotina , Química Click , Cobre/química , Dissulfetos , Proteínas/química
13.
Chembiochem ; 22(21): 3082-3089, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34387015

RESUMO

Tuberculosis is a global health problem caused by infection with the Mycobacterium tuberculosis (Mtb) bacteria. Although antibiotic treatment has dramatically reduced the impact of tuberculosis on the population, the existence and spreading of drug resistant strains urgently demands the development of new drugs that target Mtb in a different manner than currently used antibiotics. The prokaryotic ubiquitin-like protein (Pup) proteasome system is an attractive target for new drug development as it is unique to Mtb and related bacterial genera. Using a Pup-based fluorogenic substrate, we screened for inhibitors of Dop, the Mtb depupylating protease, and identified I-OMe-Tyrphostin AG538 (1) and Tyrphostin AG538 (2). The hits were validated and determined to be fast-reversible, non-ATP competitive inhibitors. We synthesized >25 analogs of 1 and 2 and show that several of the synthesized compounds also inhibit the depupylation actions of Dop on native substrate, FabD-Pup. Importantly, the pupylation activity of PafA, the sole Pup ligase in Mtb, was also inhibited by some of these compounds.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Desenvolvimento de Medicamentos , Inibidores Enzimáticos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Tirfostinas/farmacologia , Ubiquitinas/antagonistas & inibidores , Antibacterianos/síntese química , Antibacterianos/química , Proteínas de Bactérias/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Testes de Sensibilidade Microbiana , Estrutura Molecular , Mycobacterium tuberculosis/enzimologia , Relação Estrutura-Atividade , Tirfostinas/síntese química , Tirfostinas/química , Ubiquitinas/metabolismo
14.
PLoS Pathog ; 15(10): e1008086, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31658303

RESUMO

Plasmodium parasites are the causative agents of malaria, a disease with wide public health repercussions. Increasing drug resistance and the absence of a vaccine make finding new chemotherapeutic strategies imperative. Components of the ubiquitin and ubiquitin-like pathways have garnered increased attention as novel targets given their necessity to parasite survival. Understanding how these pathways are regulated in Plasmodium and identifying differences to the host is paramount to selectively interfering with parasites. Here, we focus on Nedd8 modification in Plasmodium falciparum, given its central role to cell division and DNA repair, processes critical to Plasmodium parasites given their unusual cell cycle and requirement for refined repair mechanisms. By applying a functional chemical approach, we show that deNeddylation is controlled by a different set of enzymes in the parasite versus the human host. We elucidate the molecular determinants of the unusual dual ubiquitin/Nedd8 recognition by the essential PfUCH37 enzyme and, through parasite transgenics and drug assays, determine that only its ubiquitin activity is critical to parasite survival. Our experiments reveal interesting evolutionary differences in how neddylation is controlled in higher versus lower eukaryotes, and highlight the Nedd8 pathway as worthy of further exploration for therapeutic targeting in antimalarial drug design.


Assuntos
Proteína NEDD8/metabolismo , Plasmodium falciparum/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Antimaláricos/farmacologia , Linhagem Celular , Células HEK293 , Humanos , Hidrólise , Malária Falciparum/tratamento farmacológico , Malária Falciparum/patologia , Ubiquitinação/fisiologia
15.
Chemistry ; 27(7): 2506-2512, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33075184

RESUMO

Legionnaires' disease is caused by infection with the intracellularly replicating Gram-negative bacterium Legionella pneumophila. This pathogen uses an unconventional way of ubiquitinating host proteins by generating a phosphoribosyl linkage between substrate proteins and ubiquitin by making use of an ADPribosylated ubiquitin (UbADPr ) intermediate. The family of SidE effector enzymes that catalyze this reaction is counteracted by Legionella hydrolases, which are called Dups. This unusual ubiquitination process is important for Legionella proliferation and understanding these processes on a molecular level might prove invaluable in finding new treatments. Herein, a modular approach is used for the synthesis of triazole-linked UbADPr , and analogues thereof, and their affinity towards the hydrolase DupA is determined and hydrolysis rates are compared to natively linked UbADPr . The inhibitory effects of modified Ub on the canonical eukaryotic E1-enzyme Uba1 are investigated and rationalized in the context of a high-resolution crystal structure reported herein. Finally, it is shown that synthetic UbADPr analogues can be used to effectively pull-down overexpressed DupA from cell lysate.


Assuntos
ADP-Ribosilação , Legionella pneumophila/enzimologia , Doença dos Legionários/microbiologia , Ubiquitina/química , Ubiquitina/metabolismo , Proteínas de Bactérias/metabolismo , Células HEK293 , Humanos , Hidrolases/metabolismo , Legionella pneumophila/crescimento & desenvolvimento , Enzimas Ativadoras de Ubiquitina/metabolismo , Ubiquitinação
16.
Chembiochem ; 21(20): 2903-2907, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32421893

RESUMO

Stable NAD+ analogues carrying single atom substitutions in either the furanose ring or the nicotinamide part have proven their value as inhibitors for NAD+ -consuming enzymes. To investigate the potential of such compounds to inhibit the adenosine diphosphate ribosyl (ADPr) transferase activity of the Legionella SdeC enzyme, we prepared three NAD+ analogues, namely carbanicotinamide adenosine dinucleotide (c-NAD+ ), thionicotinamide adenosine dinucleotide (S-NAD+ ) and benzamide adenosine dinucleotide (BAD). We optimized the chemical synthesis of thionicotinamide riboside and for the first time used an enzymatic approach to convert all three ribosides into the corresponding NAD+ mimics. We thus expanded the known scope of substrates for the NRK1/NMNAT1 enzyme combination by turning all three modified ribosides into NAD+ analogues in a scalable manner. We then compared the three NAD+ mimics side-by-side in a single assay for enzyme inhibition on Legionella effector enzyme SdeC. The class of SidE enzymes to which SdeC belongs was recently identified to be important in bacterial virulence, and we found SdeC to be inhibited by S-NAD+ and BAD with IC50 values of 28 and 39 µM, respectively.


Assuntos
Legionella pneumophila/enzimologia , NAD/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Modelos Moleculares , Conformação Molecular , NAD/síntese química , NAD/química , Inibidores de Poli(ADP-Ribose) Polimerases/síntese química , Inibidores de Poli(ADP-Ribose) Polimerases/química
17.
Curr Top Microbiol Immunol ; 420: 111-130, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30178261

RESUMO

Ubiquitination is a frequently occurring and very diverse posttranslational modification influencing a wide scope of cellular processes. Ubiquitin (Ub) has the unique ability to form eight different lysine-linked polymeric chains, mixed chains and engages with ubiquitin-like (Ubl) molecules. The distinct signals evoked by specific enzymes play a crucial role in, for instance, proteasome-mediated protein degradation, cell cycle regulation, and DNA damage responses. Due to the large variety of cellular functions that this posttranslational modification influences, the enzymes that construct such Ub modifications, and subsequently controle and degrade these signals, is enormous. In this chapter, we will discuss the current state-of-the-art of activity-based probes, reporter substrates, and other relevant tools based on Ub as recognition element, to study the enzymes involved in the complex system of ubiquitination.


Assuntos
Proteínas de Bactérias/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Ubiquitinação , Proteínas Virais/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Enzimas/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Proteínas Virais/antagonistas & inibidores
18.
Chembiochem ; 20(1): 62-65, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30264452

RESUMO

The enzyme-mediated construction of poly-ubiquitin (Ub) chains on target proteins leads to a variety of cellular responses and is involved in processes ranging from protein degradation to cell cycle control and immune responses. This complex post-translational modification system is under intense investigation, but generation of specific Ub chains and tools made thereof is not always trivial. We discovered that native methionine-1-linked polymeric ubiquitin chains can be constructed in a single chemical reaction. We validate correct folding and regioselective attachment of such chains using linkage specific proteases and further demonstrate that these poly-Ub chains can be converted into thioesters by the activating E1-enzyme. Subsequent ligation reactions using these in situ prepared thioesters leads to poly-ubiquitinated peptides.


Assuntos
Metionina/química , Poliubiquitina/síntese química , Polimerização , Enzimas Ativadoras de Ubiquitina/química
19.
Angew Chem Int Ed Engl ; 57(43): 14164-14168, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30188611

RESUMO

Ubiquitin-fold modifier 1 (UFM1) is a reversible post-translational modifier that is covalently attached to target proteins through an enzymatic cascade and removed by designated proteases. Abnormalities in this process, referred to as Ufmylation, have been associated with a variety of human diseases. Given this, the UFM1-specific enzymes represent potential therapeutic targets; however, understanding of their biological function has been hampered by the lack of chemical tools for activity profiling. To address this unmet need, a diversifiable platform for UFM1 activity-based probes (ABPs) utilizing a native chemical ligation (NCL) strategy was developed, enabling the generation of a variety of tools to profile both UFM1 conjugating and deconjugating enzymes. The use of the probes is demonstrated in vitro and in vivo for monitoring UFM1 enzyme reactivity, opening new research avenues.


Assuntos
Sondas Moleculares , Peptídeo Hidrolases/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Cromatografia Líquida de Alta Pressão , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Eletroporação , Células HeLa , Humanos , Proteínas/química
20.
Angew Chem Int Ed Engl ; 57(6): 1659-1662, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29215186

RESUMO

Current methods to prepare adenosine diphosphate ribosylated (ADPr) peptides are not generally applicable due to the labile nature of this post-translational modification and its incompatibility with strong acidic conditions used in standard solid-phase peptide synthesis. A general strategy is presented to prepare ADPr peptide analogues based on a copper-catalyzed click reaction between an azide-modified peptide and an alkyne-modified ADPr counterpart. The scope of this approach was expanded to proteins by preparing two ubiquitin ADPr analogues carrying the biological relevant α-glycosidic linkage. Biochemical validation using Legionella effector enzyme SdeA shows that clicked ubiquitin ADPr is well-tolerated and highlights the potential of this strategy to prepare ADPr proteins.


Assuntos
Adenosina Difosfato Ribose/química , Peptídeos/química , Proteínas/química , Triazóis/química , ADP-Ribosilação , Alcinos/química , Azidas/química , Catálise , Cobre/química , Reação de Cicloadição , Ubiquitina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA